US8758766B2 - Modified Streptococcus pneumoniae pneumolysin (PLY) polypeptides - Google Patents

Modified Streptococcus pneumoniae pneumolysin (PLY) polypeptides Download PDF

Info

Publication number
US8758766B2
US8758766B2 US13/140,894 US200913140894A US8758766B2 US 8758766 B2 US8758766 B2 US 8758766B2 US 200913140894 A US200913140894 A US 200913140894A US 8758766 B2 US8758766 B2 US 8758766B2
Authority
US
United States
Prior art keywords
plyd1
ply
wtply
mply
polypeptide
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Fee Related
Application number
US13/140,894
Other languages
English (en)
Other versions
US20110287046A1 (en
Inventor
Eliud Oloo
Raymond Oomen
Martina Ochs
Jeremy Yethon
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
KINGDOM OF NETHERLANDS REPRESENTED BY MINISTER OF HEALTH WELFARE AND SPORT ON BEHALF OF MINISTER NATIONAL INSTITUTE OF PUBLIC HEALTH AND ENVIRONMENT
Kingdom of Netherlands Represented by Mininster of Health Welfare and Sport on Behalf of Minster National Institute of Public Health and Environment
Original Assignee
Kingdom of Netherlands Represented by Mininster of Health Welfare and Sport on Behalf of Minster National Institute of Public Health and Environment
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Kingdom of Netherlands Represented by Mininster of Health Welfare and Sport on Behalf of Minster National Institute of Public Health and Environment filed Critical Kingdom of Netherlands Represented by Mininster of Health Welfare and Sport on Behalf of Minster National Institute of Public Health and Environment
Priority to US13/140,894 priority Critical patent/US8758766B2/en
Publication of US20110287046A1 publication Critical patent/US20110287046A1/en
Assigned to SANOFI PASTEUR LIMITED reassignment SANOFI PASTEUR LIMITED ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: OCHS-ONOLEMHEMHEN, MARTINA, OLOO, ELIUD, OOMEN, RAYMOND, YETHON, JEREMY
Assigned to NETHERLANDS VACCINE INSTITUTE reassignment NETHERLANDS VACCINE INSTITUTE ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: SANOFI PASTEUR LIMITED
Assigned to THE KINGDOM OF THE NETHERLANDS, REPRESENTED BY THE MINISTER OF HEALTH, WELFARE AND SPORT, ON BEHALF OF THE MINISTER, THE NATIONAL INSTITUTE OF PUBLIC HEALTH AND THE ENVIRONMENT reassignment THE KINGDOM OF THE NETHERLANDS, REPRESENTED BY THE MINISTER OF HEALTH, WELFARE AND SPORT, ON BEHALF OF THE MINISTER, THE NATIONAL INSTITUTE OF PUBLIC HEALTH AND THE ENVIRONMENT ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: NETHERLANDS VACCINE INSTITUTE
Application granted granted Critical
Publication of US8758766B2 publication Critical patent/US8758766B2/en
Expired - Fee Related legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/195Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
    • C07K14/315Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria from Streptococcus (G), e.g. Enterococci
    • C07K14/3156Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria from Streptococcus (G), e.g. Enterococci from Streptococcus pneumoniae (Pneumococcus)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/02Bacterial antigens
    • A61K39/09Lactobacillales, e.g. aerococcus, enterococcus, lactobacillus, lactococcus, streptococcus
    • A61K39/092Streptococcus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/195Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
    • C07K14/315Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria from Streptococcus (G), e.g. Enterococci
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/12Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria
    • C07K16/1267Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria from Gram-positive bacteria
    • C07K16/1275Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria from Gram-positive bacteria from Streptococcus (G)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55505Inorganic adjuvants
    • C07K2316/96
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding

Definitions

  • compositions comprising modified Streptococcus pneumoniae pneumolysin (PLY) polypeptides.
  • PLY Streptococcus pneumoniae pneumolysin
  • Nucleic acids, polypeptides encoded thereby, compositions containing the same, methods for using such nucleic acids, polypeptides and compositions are also provided.
  • Streptococcus pneumoniae is an important pathogen, causing invasive diseases such as pneumonia, meningitis, and bacteraemia. Even in regions where effective antibiotic therapy is freely available, the mortality rate from pneumococcal pneumonia can be as high as 19% in hospitalized patients. In developing countries, in excess of three million children under the age of five years die each year from pneumonia, of which S. pneumoniae is the most commonly identified causative agent. S. pneumoniae also causes less serious, but highly prevalent infections such as otitis media and sinusitis, which have a significant impact on health-care costs in developed countries. Otitis media is especially important in young children, while sinusitis affects both children and adults.
  • anti-pneumococcal conjugate vaccines have also had some well known shortcomings, including the complexity of conjugate vaccine production, which increases manufacturing costs. More important, however, is the finding that polysaccharide-based conjugate vaccines protect only against infections caused by bacteria that express the particular capsule-type represented by the vaccine. This is a problem in regions such as Latin America, Asia and Africa, in which the serotypes represented by PCV7 are responsible for much less invasive disease than elsewhere in the world (Black, et al. In: Plotkin S A, et al. eds. Vaccines, 5th Edition. WB Saunders. Chapter 23 (2008); Garcia, et al. Rev. Panam. Salud Publica.
  • Pneumolysin has been reported to be an intracellular protein that causes a variety of toxic effects in vivo when released upon lysis of pneumococci.
  • the protein is highly conserved in both amino acid sequence and antigenicity among clinical isolates, thus satisfying some basic criteria for its use as a vaccine antigen (Paton, et al. Infect. Immun. 40(2) 548-52 (1983); Lock, et al. Microb. Pathog. 5(6): 461-67 (1988)).
  • it has inherent hemolytic properties, and mutants have therefore been developed and studied for their potential as vaccines.
  • the most commonly studied pneumolysin mutant is PdB, containing a single amino acid change, Trp433Phe (Paton, et al. Infect. Immun.
  • PLY mutants including ⁇ Ala146 with a deletion of amino acid 146, and ⁇ Ala146R147, have been recently described (Kirkham, et al. Infect. Immun. 74(1): 586-93 (2006)). Both ⁇ Ala146 and ⁇ Ala146R147 were shown to lack haemolytic activity against human erythrocytes. Alum-adjuvanted ⁇ Ala146 was also shown to be as protective as alum-adjuvanted wild-type PLY.
  • PdB was found to provide a superior protection against a wide variety of strains when used in combination with other virulence factors such as PspA, PspC or PsaA (Lu, supra; Ogunniyi, supra; Berry (1995), supra). Although the PdB mutant provided significant protection in some models, it had the drawback of possessing residual hemolytic activity. However, as mentioned above, ⁇ Ala146 provided both protective immunity and a lack of haemolytic activity.
  • PlyD1 While several mutant PLY vaccines have been developed, there is a clear need in the art for additional mutants that are both protective and lack haemolytic activity.
  • PlyD1 One such mutant, PlyD1 is described herein. The data presented herein shows that PlyD1 lacks hemolytic activity, generates neutralizing antibodies that inhibit hemolysis by PLY in vitro, and is protective in certain animal models.
  • FIG. 1 Exemplary PLY amino acid sequences—alleles and modifications.
  • FIG. 2 In vitro hemolytic activity, toxin mediated lysis of sheep red blood cells, of mutations at position 293.
  • FIG. 3 Decreased lung tissue damage mediated by PLY following immunization with mPLY.
  • FIG. 4 PlyD1 protein purification based on SDS-PAGE analysis.
  • FIG. 5 Survival of CBA/J mice immunized 3 ⁇ with different doses of Adjuvanted Ply-D1.
  • mPLY modified Streptococcus pneumoniae pneumolysin proteins
  • the mPLY typically include at least one amino acid difference from wild-type PLY (“wtPLY”; e.g., any of SEQ ID NOS.: 2-42).
  • wtPLY wild-type PLY
  • mPLY induces the production of anti-wtPLY and/or anti-mPLY antibodies, which may include antibodies that prevent and/or inhibit the hemolytic activity of wtPLY and/or protective antibodies against organisms expressing wtPLY.
  • mPLY induces antibodies reactive with wtPLY and exhibits decreased hemolytic activity (including none) against erythrocytes as compared to wtPLY.
  • wtPLY is modified at one or more of amino acid 65 (threonine, “Thr65”), amino acid 293 (glycine, “Gly293”), and/or 428 (cysteine, “Cys428”) to any other amino acid.
  • wtPLY is modified at Gly293 and one or more additional amino acids such as, but not limited to, Thr65 and/or Cys428.
  • Thr65 is substituted by cysteine
  • Gly293 is substituted by cysteine
  • Cys428 is substituted by alanine.
  • nucleic acids encoding mPLY methods for making mPLY, methods for immunizing, protecting, treating, and/or preventing hosts from infection or infected by organisms expressing wtPLY, and compositions for doing the same. Other embodiments will be clear from the descriptions provided herein.
  • modified pneumolysin (PLY) polypeptides and fragments thereof are provided herein, are modified pneumolysin (PLY) polypeptides and fragments thereof, immunogenic compositions, and vaccines comprising such polypeptides and/or fragments (e.g., peptides), methods of preparing such polypeptides and/or fragments, and methods of using such polypeptides and/or fragments.
  • a modified PLY polypeptide (“mPLY”) and/or fragment of mPLY is one having differences in nucleic acid or amino acid sequence as compared to a wtPLY sequence.
  • a mPLY polypeptide typically, but not necessarily, has at least one modified amino acid sequence as compared with a wtPLY and/or fragment thereof.
  • the modification is typically an amino acid substitution, insertion, and/or deletion.
  • the mPLY polypeptide and/or fragment thereof may be preferably substantially nontoxic when compared to a protein substantially corresponding to a wtPLY protein at the same dose.
  • the modified sequences provide for changes in certain activities of mPLY that are typically associated with wtPLY, especially undesirable activities including but not limited to membrane permeation, cell lysis, and cytolytic activity against human erythrocytes and other cells (a “cell” may be human and/or non-human). It is also preferred that mPLY retain the ability to induce anti-PLY (including but not limited to anti-wtPLY) protective and/or neutralizing antibodies following administration to a host (e.g., an animal such as a mammal, e.g., human being).
  • a host e.g., an animal such as a mammal, e.g., human being.
  • such antibodies reduce (including eliminate) the hemolytic activity of wtPLY, and/or lung tissue toxicity, and/or bind to microorganisms expressing wtPLY, and/or bind to wtPLY per se, and/or provide protection against infection or dissemination of disease caused by microorganisms expressing wtPLY (e.g., Streptococcus pneumoniae ).
  • the wtPLY sequence may be any PLY expressed in an organism, including but not limited to a microorganism pathogenic (e.g., “causing disease”) in a higher organism (e.g, an animal such as a mammal, e.g., human being).
  • An exemplary pathogenic organism is Streptococcus pneumoniae .
  • Wild-type PLY polypeptides are typically encoded by a ply gene, which are highly conserved genes but do exhibit some variation (e.g., Walker, et al. Infect. Immun. 55:1184-1187 (1987); Mitchell, et al. Nucleic Acid Res. 18: 4010 (1990); Jefferies, et al. J Infect Dis. 196(6): 936-44 (2007)).
  • a PLY structural model was constructed based on sequence similarity with perfringolysin O and homology modeling using the Discovery Studio suite of programs available from Accelrys Software, Inc. The model indicates that the wt-PLY is ⁇ 20% ⁇ -helix and ⁇ 40% ⁇ -sheet with four distinct structural domains: D1 (residues 6-21, 58-147, 198-243, 319-342), D2 (residues 22-57, 343-359), D3 (residues 148-197, 244-318), and D4 (residues 360-469). Wild-type PLY may be modified in any one or more of D1, D2, D3, or D4.
  • Examplary wtPLY amino acid sequences include but are not limited to those shown in any of Walker, et al. (supra), Mitchell, et al. (supra), Jeffries, et al. (supra), FIG. 1 , SEQ ID NOS.: 2-42, GenBank Accession Nos.
  • any of the variations between these sequences may be combined with any other variations found within such wtPLY (which may include synthetic modifications described herein or elsewhere) to generate additional wtPLY that may be modified to generate mPLY.
  • Such wtPLY polypeptides typically, but not necessarily, share at least about (referring to each of the following values independently) 90%, 95%, or 99% sequence identity with the wtPLY amino acid sequence shown in SEQ ID NO.: 2 as determined using mafft (Kato, et al. Nucleic Acids Res. 30:3059-3066 (2002); Kato, et al. Brief Bioinform. 9:286-294 (2008)).
  • a wtPLY may share 98.8% sequence identity with the wtPLY sequence shown in SEQ ID NO.: 2 as determined using mafft.
  • the modifications introduced into wtPLY to produce mPLY may be made to any polypeptide sharing identity with wild-type PLY, including those polypeptides having about 90%, 95%, 98%, 99%, or 99.9% identify with SEQ ID NO.: 2. Any differences in the amino acid sequence of wtPLY polypeptides are typically but not necessarily phenotypically silent. It should be noted that the wtPLY listed herein are only exemplary; other suitable wtPLY sequences are known in the art and would be suitable to modification as described herein.
  • Exemplary mPLY polypeptides contain, for example, wtPLY modified at amino acids threonine 65 (“Thr65”), glycine 293 (“Gly293”), and/or cysteine 428 (“Cys428”).
  • Thr65 threonine 65
  • Gly293 glycine 293
  • Cys428 cysteine 428
  • a modification of any of these amino acids may be referred to using conventional nomenclature, e.g., by indicating the amino acid being modified and the modification made thereto (e.g., Thr65X, Gly293X, Cys428X, where X typically indicates the substituted amino acid, or a deletion of that amino acid).
  • wtPLY is modified at Thr65 to cysteine (“Ply-T65C”).
  • wtPLY is modified at glycine 293 to alanine (“Ply-G293A”), cysteine (“Ply-G293C”), valine (“Ply-G293V”), or threonine (“Ply-G293T”).
  • wtPLY is modified at Cys428 to alanine (“Ply-Cys428A”).
  • wtPLY is modified at Gly293 and one or more additional amino acids such as, but not limited to, Thr65 and/or Cys428.
  • wtPLY is modified at Thr65 and one or more additional amino acids such as, but not limited to, Gly293 and/or Cys428.
  • wtPLY is modified at Cys428 and one or more additional amino acids such as, but not limited to, Thr65 and/or Gly293. Modifications at Thr65, Gly293, and/or Cys428 may be made to any wtPLY to produce mPLY polypeptides. Preferred substitutions in each of these embodiments is cysteine for Thr65; cysteine, valine, or threonine for Gly293; and/or alanine for Cys428. Exemplary mPLY polypeptides are shown in SEQ ID NOS. 43 and 44. Any of these modifications may be introduced into any wtPLY described herein or elsewhere combined with any other modifications to produce mPLY polypeptides using standard molecular biology and biochemical techniques.
  • PLY (wtPLY or mPLY) activity may be assayed and characterized by methods known to those of skill in the art (e.g., Paton, et al., supra; Nato, et al. Infect Immun. 59(12): 4641-4646 (1991)). Any one or more of the assays described herein, or any other one or more suitable assays, may be used to determine the suitability of mPLY as a vaccine.
  • Two suitable, exemplary assays are the in vitro hemolytic assay and the hemolysis inhibition assay.
  • the in vitro hemolysis assay measures the hemolytic (e.g., cytolytic) activity of mPLY relative to wtPLY.
  • the hemolysis inhibition assay measures the ability of antisera raised against mPLY to inhibit hemolysis by PLY, and (typically) comparing anti-mPLY antisera to anti-wtPLY antisera. Either or both of these assays may be utilized to determine the activity of a particular mPLY. In vivo assays may also be used, including but not limited to the sepsis model, the focal pneumonia model, and the intranasal challenge models essentially as described in Example 8. Thus, a suitable mPLY may be one that exhibits lower hemolytic activity than wtPLY (e.g., via an in vitro hemolysis assay).
  • a suitable mPLY may also be one that, following administration to a host, causes the host to produce antibodies that inhibit hemolysis by wtPLY (e.g., via a hemolysis inhibition assay), is immunogenic (e.g., an immunogenic composition, one that induces the production of antibodies or a cytotoxic T cell response against wtPLY or microorganisms expressing wtPLY), protective (e.g., a prophylactic or therapeutic vaccine composition, one that induces an immune response that protects the host against infection by or limits an already-existing infection, respectively, by an organism expressing wtPLY), provides benefits as measured by any one or more of the in vivo assays described herein (e.g., sepsis model, focal pneumoniae model, intranasal challenge, lung damage assay), and/or increases or decreases expression of cytokines by macrophages as compared to wtPLY. It is to be understood that these methods for identifying and/or characterizing mPLY are
  • test polypeptides e.g., wtPLY, mPLY
  • test polypeptides may be separately serially diluted across a plastic microtiter plate in serial dilutions (e.g., 2-fold) with a highest concentration typically being about 0.5 mg/mL of test polypeptide.
  • Bovine serum albumin BSA
  • Sheep red blood cells may then be added to all wells, and the plates incubated for a sufficient period of time (e.g., 30 min.).
  • Positive controls e.g., 100% lysis measurement is obtained by the addition of 1% Trition X-100 to particular wells
  • negative controls e.g., PBS alone
  • the plate may then be centrifuged to separate the intact cells from the lysed cells.
  • the supernatant containing the lysed cells may then be transferred to a fresh plate and subjected to an A 540 hemoglobin release assay (lysed cells release hemoglobin).
  • the specific activity may be determined as the inverse of protein concentration (mg/mL) at which 50% hemolysis occurred relative to the positive control.
  • mPLT may have a specific activity of about 0%, 0.0005%, 0.001%, 0.005%, 0.01%, 0.05%, 0.1%, 0.5%, 1%, 5% or ⁇ 10% the specific activity of wtPLY.
  • a specific activity of approximately within these ranges typically indicates the particular mPLY is within acceptable parameters.
  • anti-PLY wtPLY or mPLY
  • wtPLY or mPLY antisera is produced essentially as described in the art or herein and serially diluted (e.g., two-fold) across a microtiter plate (e.g., a 96-well plate) and a constant amount of wtPLY is added to each well to be analyzed.
  • Sheep red blood cells are added to all wells and the assay proceeds as per the in vitro hemolysis assay described in the preceding paragraph.
  • the data are plotted as percent hemolysis versus antiserum titer (log scale).
  • the 50% hemolysis inhibition titer is taken as the inverse of the serum dilution at which the percent hemolysis is reduced to 50%.
  • a particular mPLY have a similar (e.g., anywhere from about 75 to about 100%) or higher 50% hemolysis inhibition titer than wtPLY.
  • a particular mPLY induce an antisera having a 50% hemolysis inhibition titer (using serial two-fold dilutions) of about 1:16, 1:32, 1:64, 1:128, 1:256, 1:512, 1:1024, 1:2048, 1:5096, or 1:10,192, or greater.
  • a 50% hemolysis inhibition titer of approximately within these ranges typically indicates the particular mPLY is within acceptable parameters.
  • the mPLY may have both an acceptable specific activity as determined by the in vitro hemolysis assay and an acceptable 50% hemolysis inhibition titer.
  • a suitable mPLY may have a specific activity (as determined using the in vitro hemolysis assay) of about (referring to each of the following values independently) 0%, 0.0005%, 0.001%, 0.005%, 0.01%, 0.05%, 0.1%, 0.5%, 1%, 5% or ⁇ 10% the specific activity of wtPLY, and a 50% hemolysis inhibition titer (using serial two-fold dilutions) of about 1:128, 1:256, 1:512, 1:1024, 1:2048, 1:5096, or 1:10,192.
  • wtPLY is known to activate macrophages to generate cytokines (Malley, et al. PNAS USA, 100(4): 1966-71 (2003)). Suitable mPLY may be identified by determining macrophage activation by measuring mPly-induced cytokine production (e.g., (IL-1 ⁇ , IL-6 and IL-10) in vitro. Macrophage-like cells (e.g., human MM6 and mouse J774A.1 cells) may be incubated overnight with wtPLY or mPLY, and treated or not treated with proteinase K and heat (to distinguish false positives due to lipopolysaccharide (LPS) contamination in the treated group).
  • mPly-induced cytokine production e.g., (IL-1 ⁇ , IL-6 and IL-10
  • Macrophage-like cells e.g., human MM6 and mouse J774A.1 cells
  • LPS lipopolysaccharide
  • Cytokine production (e.g., IL-6, IL-10, TNF- ⁇ , IL-1 ⁇ ) may be measured by ELISA after overnight incubation.
  • a suitable mPLY will induce expression of cytokines (e.g., IL-1 ⁇ , IL-6 and IL-10) more than will wtPLY.
  • a suitable mPLY will induce expression of cytokines (e.g., IL-1 ⁇ , IL-6 and IL-10) less than will wtPLY.
  • the difference in the levels of expression induced by mPLY and wtPLY may be, for instance, about 10, 20, 30, 40, 50, 60, 70, 80, 90, 100% or more than 100%.
  • mice may be immunized with mPLY (e.g., a dose of about 1, 2, 5 or 10 ⁇ g for a sufficient period of time), and then challenged with wtPLY (e.g., a dose of about 1, 2, 5, or 10 ⁇ g for a sufficient period of time) or an organism expressing wtPLY. After a sufficient period of time (e.g., a period of hours, days, weeks), the lungs may be harvested and stained to observe tissue damage.
  • mPLY e.g., a dose of about 1, 2, 5 or 10 ⁇ g for a sufficient period of time
  • wtPLY e.g., a dose of about 1, 2, 5, or 10 ⁇ g for a sufficient period of time
  • the lungs may be harvested and stained to observe tissue damage.
  • wtPLY typically causes perivascular edema, thickened, disrupted alveolar walls, diminished alveolar space, and fluid and blood infiltration of the alveolar spaces.
  • a suitable mPLY may induce antibodies that reduce lung damage by, for instance, about 10, 20, 30, 40, 50, 60, 70, 80, 90, or 100% as compared to that induced by wtPLY or an organism expressing wtPLY.
  • a suitable mPLY exhibit immunogenic properties (e.g., inducing a neutralizing and/or protective immune response following administration to a host).
  • the presence of neutralizing and/or protective immune response may be demonstrated by demonstrating that infection by organisms expressing wtPLY is decreased in immunized individuals (e.g., human beings, or using animal models).
  • Suitable animal models include the sepsis model, the focal pneumoniae model, and the intranasal challenge model.
  • test animals e.g., mice or similar model
  • mPLY with or without one or more adjuvants at suitable timepoints such as days 0, 7, 14, 21, 28, 35, 42
  • a suitable amount of time e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 weeks
  • wt-PLY expressing organism e.g., 10 7 colony forming units (cfu) of S. pneumoniae serotype 6B.
  • mice may be monitored for mortality, and sample bleeds may be drawn.
  • Sera may be analyzed for total PLY-specific IgG response using, for example, an antibody ELISA and for PLY neutralizing capacity using, for example, an inhibition of hemolysis assay.
  • Statistical analysis e.g., Fisher's exact test, Wilcoxon test, Mann-Whitney Test
  • mPlyD1 may also be tested using a focal pneumonia mouse model. Briefly, animals may be immunized (e.g., subcutaneously, intravenously, intramuscularly, intradermally, intranodally, intranasally) with a purified recombinant PlyD1 protein with or without adjuvant.
  • Test animals e.g., mice or similar model
  • mice may be immunized with mPLY (with or without one or more adjuvants at suitable timepoints such as days 0, 7, 14, 21, 28, 35, 42), and then after a suitable amount of time (e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 weeks) challenged by intranasally (IN) with a suitable amount of wt-PLY expressing organism (e.g., 3 ⁇ 7 ⁇ 10 6 cfu of S. pneumoniae strain EF3030 (serotype 19F)). Following the challenge, mice may be monitored for mortality, and sample bleeds may be drawn.
  • suitable time e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 weeks
  • wt-PLY expressing organism e.g., 3 ⁇ 7 ⁇ 10 6 cfu of S. pneumoniae strain EF3030 (serotype 19F)
  • Sera may be analyzed for total PLY-specific IgG response using, for example, an antibody ELISA and for PLY neutralizing capacity using, for example, an inhibition of hemolysis assay.
  • Statistical analysis e.g., Fisher's exact test, Wilcoxon test, Mann-Whitney Test
  • mice may be immunized (e.g., subcutaneously, intravenously, intramuscularly, intradermally, intranodally, intranasally) with purified recombinant PlyD1 proteins at an effective dose (e.g., from about 0.25 to 25 ⁇ g) with or without one or more adjuvants at suitable timepoints such as days 0, 7, 14, 21, 28, 35, 42), and then after a suitable amount of time (e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 weeks) challenged by intranasally (IN) with a suitable amount of wt-PLY expressing organism (e.g., a lethal dose (5 ⁇ 10 5 cfu) of S.
  • an effective dose e.g., from about 0.25 to 25 ⁇ g
  • suitable time e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 weeks
  • wt-PLY expressing organism e.g., a lethal dose (5 ⁇ 10 5 cfu) of
  • mice may be monitored for mortality, and sample bleeds may be drawn.
  • Sera may be analyzed for total PLY-specific IgG response using, for example, an antibody ELISA and for PLY neutralizing capacity using, for example, an inhibition of hemolysis assay.
  • Statistical analysis e.g., Fisher's exact test, Wilcoxon test, Mann-Whitney Test
  • Immunogenic compositions and vaccines containing mPLY polypeptides and/or fragments thereof may be used to treat diseases such as, for example, invasive pneumococcal such as pneumonia, meningitis, otitis media, and bacteremia.
  • the polypeptides and fragments exhibit substantially reduced toxicity compared to the native toxin.
  • Nucleic acid sequences encoding the mPLY polypeptides and/or fragments, vectors containing such nucleic acid sequences, and host cells capable of expressing the mutant PLY polypeptides and/or fragments are also provided.
  • the mPLY polypeptides and/or fragments preferably exhibit decreased toxicity relative to wtPLY (e.g., lower hemolytic/cytolytic activity) and elicit neutralizing antibodies which are cross-reactive with antibodies elicited by the wtPLY.
  • mPLY polypeptides and/or fragments may be used to produce immunological compositions which may include vaccines.
  • An immunological composition is one that, upon administration to a host (e.g., an animal), induces or enhances an immune response directed against the antigen or immunogen contained within the composition. This response may include the generation of antibodies (e.g, through the stimulation of B cells) or a T cell-based response (e.g., a cytolytic response).
  • a protective or neutralizing immune response is one that is detrimental to the infectious organism corresponding to the antigen (e.g, from which the antigen was derived) and beneficial to the host (e.g., by reducing or preventing infection).
  • protective or neutralizing antibodies and/or cellular responses may be reactive to mPLY, wtPLY, or fragments thereof and reduce or inhibit the lethality of wtPLY (or organisms expressing wtPLY or mPLY) when tested in animals.
  • An immunological composition that, upon administration to a host, results in a protective or neutralizing immune response, may be considered a vaccine.
  • Immunological compositions comprising one or more mPLY polypeptides may also include one or more additional antigens, such as one or more antigens of S. pneumoniae .
  • additional antigens include, for example, PcPA and/or PhtD.
  • Other variations of immunological compositions are also contemplated as would be understood by one of skill in the art.
  • mPLY polypeptides and/or fragments thereof typically have at least one nucleic acid and/or one amino acid substitution. Modified polypeptides may also exhibit at least one change in a biological function (e.g., immunogenicity, haemolytic activity) compared with a wtPLY. mPLY polypeptides or fragments thereof are preferably substantially nontoxic when compared to a wtPLY protein at the same dose, elicit antibodies that are preferably protective or neutralizing and cross-reactive with antibodies elicited by a wtPLY protein.
  • a biological function e.g., immunogenicity, haemolytic activity
  • a mPLY polypeptide and/or fragment thereof may be generated using a variety of methods including, but not limited to, site-directed mutagenesis, random mutagenesis, conventional mutagenesis, in vitro mutagenesis, spontaneous mutagenesis and chemical synthesis. Methods of mutagenesis can be found in Sambrook et al., A Guide to Molecular Cloning, Cold Spring Harbour, N.Y. (1989) and Sambrook and Russel. Molecular Cloning: A Laboratory Manual (2001), for instance.
  • This disclosure further relates to antibodies, preferably protective or neutralizing antibodies, generated using a mPLY polypeptide or fragment thereof where the resultant antibodies are reactive to wtPLY and/or fragments thereof. Also provided are methods for eliciting the production of antibodies, which may be protective and/or neutralizing, reactive to the mPLY or fragments thereof. The antibodies may elicit both active and passive immunity. The mPLY polypeptides and/or fragments thereof may also be used to identify and isolate antibodies, which may be protective and/or neutralizing, that are cross-reactive with those elicited by wtPLY.
  • Nucleic acids encoding mPLY polypeptides are also provided. Also provided are variants of such sequences, including degenerate variants thereof.
  • a nucleic acid molecule encoding the mPLY polypeptide and/or fragment may be inserted into one or more expression vectors, as discussed below in greater detail.
  • the mPLY polypeptide and/or fragment is/are encoded by nucleotides corresponding to the amino acid sequence.
  • the particular combinations of nucleotides that encode the various amino acids are well known in the art, as described in various references used by those skilled in the art (i.e., Lewin, B. Genes V , Oxford University Press, 1994), and as shown in Table 1 below.
  • Nucleic acid variants may use any combination of nucleotides that encode the polypeptide of interest.
  • Modified PLY polypeptides and/or fragments are typically selected to include least one amino acid substitution, and optionally at least one change in a biological function compared with wtPLY.
  • Modified PLY proteins or fragment thereof may be selected to ensure that they are substantially non-toxic when compared to a protein substantially corresponding to a wtPLY protein at the same dose and elicit neutralizing antibodies which may be cross-reactive with antibodies elicited by a wtPLY protein.
  • mPLY polypeptides and fragments thereof may be useful in immunogenic compositions or vaccines against invasive pneumococcal disease (e.g., pneumonia, meningitis, otitis media, bacteremia) caused by Streptococcus pneumoniae.
  • invasive pneumococcal disease e.g., pneumonia, meningitis, otitis media, bacteremia
  • Amino acid substitution may be conservative or non-conservative. Substitutions that result in a structural change that may affect mPLY activity include the following:
  • Conservative amino acid substitutions may involve a substitution of a native amino acid residue with a non-native residue such that there is little or no effect on the size, polarity, charge, hydrophobicity, or hydrophilicity of the amino acid residue at that position and, in particular, does not result in decreased immunogenicity. Suitable conservative amino acid substitutions are shown in Table 2.
  • nucleic acids encoding mPLY polypeptides and/or fragments substituted may be selected based on the degeneracy of the genetic code.
  • the nucleic acid is a recombinant DNA molecule useful for expressing a polypeptide in a cell (e.g., an expression vector)
  • such a substation may result in the expression of a polypeptide with the same amino acid sequence as that originally encoded by the DNA molecule.
  • substitutions may be conservative, or non-conservative, or any combination thereof.
  • Suitable variants of mPLY polypeptides and/or fragments provided herein using well-known techniques. For identifying suitable areas of the molecule that may be changed without destroying biological activity (i.e., pore-forming activity, red blood cell (RBC) agglutination, RBC hemolysis, MHC binding, immunogenicity), one skilled in the art may target areas not believed to be important for that activity. For example, when PLY derivatives with similar activities from the same species or from other species are known, one skilled in the art may compare the amino acid sequence of a polypeptide to such similar polypeptides. By performing such analyses, one can identify residues and portions of the molecules that are conserved.
  • biological activity i.e., pore-forming activity, red blood cell (RBC) agglutination, RBC hemolysis, MHC binding, immunogenicity
  • the mPLY polypeptides and/or fragments described herein may include fusion polypeptide segments that assist in purification or detection of the polypeptides. Fusions can be made either at the amino terminus or at the carboxy terminus of the subject polypeptide variant thereof. Fusions may be direct with no linker or adapter molecule or may be through a linker or adapter molecule. A linker or adapter molecule may be one or more amino acid residues, typically from about 2 to about 50 amino acid residues. A linker or adapter molecule may also be designed with a cleavage site for a DNA restriction endonuclease or for a protease to allow for the separation of the fused moieties.
  • fusion polypeptides can be derivatized according to the methods described herein.
  • Suitable fusion segments include, among others, metal binding domains (e.g., a poly-histidine segment), immunoglobulin binding domains (i.e., Protein A, Protein G, T cell, B cell, Fc receptor, or complement protein antibody-binding domains), sugar binding domains (e.g., a maltose binding domain), and/or a “tag” domain (i.e., at least a portion of ⁇ -galactosidase, a strepavidin-derived tag peptide, a T7 tag peptide, a FLAG peptide, or other peptides that can be purified using compounds that bind to the domain, such as monoclonal antibodies).
  • metal binding domains e.g., a poly-histidine segment
  • immunoglobulin binding domains i.e., Protein A, Protein G, T cell, B cell, Fc receptor, or complement
  • This tag is typically fused to the polypeptide upon expression of the polypeptide, and can serve as a means for affinity purification of the sequence of interest polypeptide from the host cell. Affinity purification can be accomplished, for example, by column chromatography using antibodies against the tag as an affinity matrix.
  • affinity purification can be accomplished, for example, by column chromatography using antibodies against the tag as an affinity matrix.
  • the tag can subsequently be removed from the purified sequence of interest polypeptide by various means such as using certain peptidases for cleavage.
  • the mPLY polypeptides and/or fragments may be directly or indirectly (i.e., using an antibody) labeled or tagged in a manner which enables it to be detected.
  • Labels include fluorochromes such as fluorescein, rhodamine, phycoerythrin, Europium and Texas Red, chromogenic dyes such as diaminobenzidine, radioisotopes, macromolecular colloidal particles or particulate material such as latex beads that are coloured, magnetic or paramagnetic, binding agents such as biotin and digoxigenin, and biologically or chemically active agents that can directly or indirectly cause detectable signals to be visually observed, electronically detected or otherwise recorded, for example in a FACS, ELISA, Western blot, TRFIA, immunohistochemistry, evanescence, Luminex bead array, or dipstick or other lateral flow assay format.
  • Suitable antibody-binding molecules for use in such methods may include immunoglobulin-binding antibodies, for example anti-human Ig antibodies, anti-human Ig antibodies, anti-human antibodies specific for Ig isotypes or for subclasses of IgG, or specific for Staphylococcal protein A or G.
  • immunoglobulin-binding antibodies for example anti-human Ig antibodies, anti-human Ig antibodies, anti-human antibodies specific for Ig isotypes or for subclasses of IgG, or specific for Staphylococcal protein A or G.
  • Preferred fluorescent tag proteins include those derived from the jelly fish protein known as green fluorescent protein (GFP). Further information on GFP and other fluorophores is given in the following publications: Tsien R Y, “The Green Fluorescent Protein” Annual Reviews of Biochemistry 1998; 67:509-544 Verkhusha, V and Lukyanov, K. “The Molecular Properties and Applications of Anthoza Fluorescent Proteins and Chromophores” Nature Biotechnology 2004; 22:289-296. Plasmid vectors encoding a wide range of fluorescent tag proteins are commercially available from various suppliers including an array of “Living Colours™ Fluorescent Proteins” available commercially from Clontech Laboratories, Inc.
  • Suitable fluorescent proteins derived from GFP are the red-shifted variant EGFP, the cyan shifted variant ECFP and the yellow shifted variant EYFP.
  • EGFP is preferred as the fluorescent marker because it gives bright fluorescence combined with minimal effect on the antigenic properties of the target antigen.
  • Alternative fluorescent marker proteins are commercially available.
  • Biologically or chemically active agents include enzymes, which catalyse reactions that develop or change colours or cause changes in electrical properties, for example, and may also be utilized. They may be molecularly excitable, such that electronic transitions between energy states result in characteristic spectral absorptions or emissions. They may include chemical entities used in conjunction with biosensors.
  • Biotin/avidin or biotin/streptavidin and alkaline phosphatase detection systems may be employed. Further examples include horseradish peroxidase and chemiluminescence
  • the non-immobilized, antibody-binding molecule, or polypeptide may be detected using an antibody which binds to said non-immobilised antibody-binding molecule or polypeptide.
  • a suitable detection antibody may be labeled by means of fluorescence.
  • the label may be a fluorescent marker (tag) which is used to label the target antigen directly such that the antigen and the fluorescent marker form a fusion protein.
  • the antigen may be labeled with the tag bound to those antibodies, and the complex formed thereby detected using immunoprecipitation.
  • the fluorescence associated with the tag may then be used to determine that protein has been precipitated (qualitative determination) or to determine the amount of protein precipitated (quantitative determination).
  • soluble extracts of a fluorescence-tagged antigen may be incubated with patient sera for an appropriate period of time such as overnight at 4° C. (typically 10-15 ⁇ l of serum to 300-500 ⁇ l of extract or less) to allow antibodies to bind to the antigen.
  • Protein A or Protein G Sepharose beads preincubated with low IgG fetal calf serum (Sigma) to block non-specific binding, are then added to the extract/serum mix containing the tagged protein/antibody complexes, and mixed with gentle rotation for 1 to 2 hours at room temperature.
  • the antibodies within the serum including those that specifically bind the tagged protein, are bound by the protein A/G beads.
  • the protein A/G Sepharose beads are then washed in a suitable buffer (typically 10 mM Tris-HCl pH 7.4, 100 mM NaCl/ImM EDTA/1% Triton X-100) to remove any unbound tagged protein.
  • the assays described herein for detecting antibodies immunoreactive with streptococcal antigens may also be combined with other assays useful for detecting streptococcal infection.
  • these assays i.e., ELISA
  • PCR polymerase chain reaction
  • an ELISA assay may be combined with an immunoprecipitation assay.
  • a PCR-based assay may be combined with an immunoprecipitation assay. Combining the various assays described herein may serve to even further increase the sensitivity of detection and further decrease the negative predictive value of the data.
  • expression vectors may also be suitable for use.
  • Expression vectors are typically comprised of a flanking sequence operably linked to a heterologous nucleic acid sequence encoding a polypeptide (the “coding sequence”).
  • a flanking sequence is preferably capable of effecting the replication, transcription and/or translation of the coding sequence and is operably linked to a coding sequence.
  • operably linked indicates that the nucleic acid sequences are configured so as to perform their usual function.
  • a promoter is operably linked to a coding sequence when the promoter is capable of directing transcription of that coding sequence.
  • a flanking sequence need not be contiguous with the coding sequence, so long as it functions correctly.
  • intervening untranslated yet transcribed sequences can be present between a promoter sequence and the coding sequence and the promoter sequence can still be considered operably linked to the coding sequence.
  • Flanking sequences may be homologous (i.e., from the same species and/or strain as the host cell), heterologous (i.e., from a species other than the host cell species or strain), hybrid (i.e., a combination of flanking sequences from more than one source), or synthetic.
  • a flanking sequence may also be a sequence that normally functions to regulate expression of the nucleotide sequence encoding the polypeptide in the genome of the host may also be utilized.
  • the flanking sequence is a transcriptional regulatory region that drives high-level gene expression in the target cell.
  • the transcriptional regulatory region may comprise, for example, a promoter, enhancer, silencer, repressor element, or combinations thereof.
  • the transcriptional regulatory region may be either constitutive or tissue- or cell-type specific (i.e., the region drives higher levels of transcription in one type of tissue or cell as compared to another).
  • the source of a transcriptional regulatory region may be any prokaryotic or eukaryotic organism, any vertebrate or invertebrate organism, or any plant, provided that the flanking sequence is functional in, and can be activated by, the host cell machinery.
  • a wide variety of transcriptional regulatory regions may be utilized.
  • Suitable transcriptional regulatory regions include, among others, the CMV promoter (i.e., the CMV-immediate early promoter); promoters from eukaryotic genes (i.e., the estrogen-inducible chicken ovalbumin gene, the interferon genes, the gluco-corticoid-inducible tyrosine aminotransferase gene, and the thymidine kinase gene); and the major early and late adenovirus gene promoters; the SV40 early promoter region (Bernoist and Chambon, 1981, Nature 290:304-10); the promoter contained in the 3′ long terminal repeat (LTR) of Rous sarcoma virus (RSV) (Yamamoto, et al., 1980, Cell 22:787-97); the herpes simplex virus thymidine kinase (HSV-TK) promoter (Wagner et al., 1981, Proc.
  • the CMV promoter i.e., the
  • Tissue- and/or cell-type specific transcriptional control regions include, for example, the elastase I gene control region which is active in pancreatic acinar cells (Swift et al., 1984, Cell 38:639-46; Ornitz et al., 1986, Cold Spring Harbor Symp. Quant. Biol.
  • the nucleic acid molecule may be administered as part of a viral and non-viral vector.
  • a DNA vector is utilized to deliver nucleic acids encoding the targeted immunogen and/or associated molecules (i.e., co-stimulatory molecules, cytokines or chemokines) to the patient.
  • various strategies may be utilized to improve the efficiency of such mechanisms including, for example, the use of self-replicating viral replicons (Caley, et al. 1999. Vaccine, 17: 3124-2135; Dubensky, et al. 2000. Mol. Med. 6: 723-732; Leitner, et al. 2000. Cancer Res. 60: 51-55), codon optimization (Liu, et al. 2000.
  • Vaccine, 19: 2647-2656 incorporation of stimulatory motifs such as CpG (Gurunathan, supra; Leitner, supra), sequences for targeting of the endocytic or ubiquitin-processing pathways (Thomson, et al. 1998. J. Tirol. 72: 2246-2252; Velders, et al. 2001. J. Immunol. 166: 5366-5373), prime-boost regimens (Gurunathan, supra; Sullivan, et al. 2000. Nature, 408: 605-609; Hanke, et al. 1998. Vaccine, 16: 439-445; Amara, et al. 2001.
  • stimulatory motifs such as CpG (Gurunathan, supra; Leitner, supra), sequences for targeting of the endocytic or ubiquitin-processing pathways (Thomson, et al. 1998. J. Tirol. 72: 2246-2252; Velders, et al. 2001. J
  • viral vectors that have been successfully utilized for introducing a nucleic acid to a host include retrovirus, adenovirus, adeno-associated virus (AAV), herpes virus, and poxvirus, among others.
  • the vectors may be constructed using standard recombinant techniques widely available to one skilled in the art. Such techniques may be found in common molecular biology references such as Molecular Cloning: A Laboratory Manual (Sambrook, et al., 1989, Cold Spring Harbor Laboratory Press), Gene Expression Technology (Methods in Enzymology, Vol. 185, edited by D. Goeddel, 1991. Academic Press, San Diego, Calif.), and PCR Protocols: A Guide to Methods and Applications (Innis, et al. 1990. Academic Press, San Diego, Calif.).
  • retroviral vectors are derivatives of lentivirus as well as derivatives of murine or avian retroviruses.
  • suitable retroviral vectors include, for example, Moloney murine leukemia virus (MoMuLV), Harvey murine sarcoma virus (HaMuSV), murine mammary tumor virus (MuMTV), SIV, BIV, HIV and Rous Sarcoma Virus (RSV).
  • MoMuLV Moloney murine leukemia virus
  • HaMuSV Harvey murine sarcoma virus
  • MuMTV murine mammary tumor virus
  • SIV BIV
  • HIV Rous Sarcoma Virus
  • retroviral vectors can incorporate multiple exogenous nucleic acid sequences. As recombinant retroviruses are defective, they require assistance in order to produce infectious vector particles. This assistance can be provided by, for example, helper cell lines encoding retrovirus structural genes.
  • Suitable helper cell lines include ⁇ 2, PA317 and PA12, among others.
  • the vector virions produced using such cell lines may then be used to infect a tissue cell line, such as NM 3T3 cells, to produce large quantities of chimeric retroviral virions.
  • Retroviral vectors may be administered by traditional methods (i.e., injection) or by implantation of a “producer cell line” in proximity to the target cell population (Culver, K., et al., 1994, Hum. Gene Ther., 5 (3): 343-79; Culver, K., et al., Cold Spring Harb. Symp. Quant. Biol., 59: 685-90); Oldfield, E., 1993, Hum.
  • the producer cell line is engineered to produce a viral vector and releases viral particles in the vicinity of the target cell. A portion of the released viral particles contact the target cells and infect those cells, thus delivering a nucleic acid to the target cell. Following infection of the target cell, expression of the nucleic acid of the vector occurs.
  • Adenoviral vectors have proven especially useful for gene transfer into eukaryotic cells (Rosenfeld, M., et al., 1991 , Science, 252 (5004): 431-3; Crystal, R., et al., 1994, Nat. Genet., 8 (1): 42-51), the study of eukaryotic gene expression (Levrero, M., et al., 1991, Gene, 101 (2): 195-202), vaccine development (Graham, F. and Prevec, L., 1992, Biotechnology, 20: 363-90), and in animal models (Stratford-Perricaudet, L., et al., 1992, Bone Marrow Transplant., 9 (Suppl.
  • Adeno-associated virus demonstrates high-level infectivity, broad host range and specificity in integrating into the host cell genome (Hermonat, P., et al., 1984, Proc. Natl. Acad. Sci. U.S.A., 81 (20): 6466-70).
  • Herpes Simplex Virus type-1 HSV-1
  • HSV-1 Herpes Simplex Virus type-1
  • Poxvirus is another useful expression vector (Smith, et al. 1983, Gene, 25 (1): 21-8; Moss, et al, 1992, Biotechnology, 20: 345-62; Moss, et al, 1992, Curr. Top. Microbiol. Immunol., 158: 25-38; Moss, et al. 1991. Science, 252: 1662-1667).
  • Poxviruses shown to be useful include vaccinia, NYVAC, avipox, fowlpox, canarypox, ALVAC, and ALVAC(2), among others.
  • NYVAC (vP866) was derived from the Copenhagen vaccine strain of vaccinia virus by deleting six nonessential regions of the genome encoding known or potential virulence factors (see, for example, U.S. Pat. Nos. 5,364,773 and 5,494,807). The deletion loci were also engineered as recipient loci for the insertion of foreign genes.
  • the deleted regions are: thymidine kinase gene (TK; J2R) vP410; hemorrhagic region (u; B13R+B14R) vP553; A type inclusion body region (ATI; A26L) vP618; hemagglutinin gene (HA; A56R) vP723; host range gene region (C7L-K1L) vP804; and, large subunit, ribonucleotide reductase (I4L) vP866.
  • NYVAC is a genetically engineered vaccinia virus strain that was generated by the specific deletion of eighteen open reading frames encoding gene products associated with virulence and host range.
  • NYVAC has been show to be useful for expressing TAs (see, for example, U.S. Pat. No. 6,265,189).
  • NYVAC (vP866), vP994, vCP205, vCP1433, placZH6H4Lreverse, pMPC6H6K3E3 and pC3H6FHVB were also deposited with the ATCC under the terms of the Budapest Treaty, accession numbers VR-2559, VR-2558, VR-2557, VR-2556, ATCC-97913, ATCC-97912, and ATCC-97914, respectively.
  • ALVAC-based recombinant viruses i.e., ALVAC-1 and ALVAC-2 are also suitable for use (see, for example, U.S. Pat. No. 5,756,103).
  • ALVAC(2) is identical to ALVAC(1) except that ALVAC(2) genome comprises the vaccinia E3L and K3L genes under the control of vaccinia promoters (U.S. Pat. No. 6,130,066; Beattie et al., 1995a, 1995b, 1991; Chang et al., 1992; Davies et al., 1993).
  • ALVAC(1) and ALVAC(2) have been demonstrated to be useful in expressing foreign DNA sequences, such as TAs (Tartaglia et al., 1993 a,b; U.S. Pat. No. 5,833,975).
  • ALVAC was deposited under the terms of the Budapest Treaty with the American Type Culture Collection (ATCC), 10801 University Boulevard, Manassas, Va. 20110-2209, USA, ATCC accession number VR-2547.
  • TROVAC refers to an attenuated fowlpox that was a plaque-cloned isolate derived from the FP-1 vaccine strain of fowlpoxvirus which is licensed for vaccination of 1 day old chicks. TROVAC was likewise deposited under the terms of the Budapest Treaty with the ATCC, accession number 2553.
  • Non-viral plasmid vectors may also be suitable in certain embodiments.
  • Preferred plasmid vectors are compatible with bacterial, insect, and/or mammalian host cells.
  • Such vectors include, for example, PCR-II, pCR3, and pcDNA3.1 (Invitrogen, San Diego, Calif.), pBSII (Stratagene, La Jolla, Calif.), pET15 (Novagen, Madison, Wiss.), pGEX (Pharmacia Biotech, Piscataway, N.J.), pEGFP-N2 (Clontech, Palo Alto, Calif.), pETL (BlueBacII, Invitrogen), pDSR-alpha (PCT pub.
  • vectors include, for example, Shigella, Salmonella, Vibrio cholerae, Lactobacillus, Bacille calmette conditioningn (BCG), and Streptococcus (see for example, WO 88/6626; WO 90/0594; WO 91/13157; WO 92/1796; and WO 92/21376).
  • BCG Bacille calmette conditioning
  • Streptococcus see for example, WO 88/6626; WO 90/0594; WO 91/13157; WO 92/1796; and WO 92/21376).
  • Many other non-viral plasmid expression vectors and systems are known in the art and may be used.
  • colloidal dispersion systems include macromolecule complexes, nanocapsules, microspheres, beads, and lipid-based systems including oil-in-water emulsions, micelles, mixed micelles, and liposomes.
  • the preferred colloidal system is a liposome, which are artificial membrane vesicles useful as delivery vehicles in vitro and in vivo.
  • RNA, DNA and intact virions can be encapsulated within the aqueous interior and be delivered to cells in a biologically active form (Fraley, R., et al., 1981, Trends Biochem. Sci., 6: 77).
  • the composition of the liposome is usually a combination of phospholipids, particularly high-phase-transition-temperature phospholipids, usually in combination with steroids, especially cholesterol. Other phospholipids or other lipids may also be used.
  • the physical characteristics of liposomes depend on pH, ionic strength, and the presence of divalent cations.
  • lipids useful in liposome production include phosphatidyl compounds, such as phosphatidylglycerol, phosphatidylcholine, phosphatidylserine, phosphatidylethanolamine, sphingolipids, cerebrosides, and gangliosides. Particularly useful are diacylphosphatidylglycerols, where the lipid moiety contains from 14-18 carbon atoms, particularly from 16-18 carbon atoms, and is saturated.
  • Illustrative phospholipids include egg phosphatidylcholine, dipalmitoylphosphatidylcholine and distearoylphosphatidylcholine.
  • a cultured cell comprising the vector is also provided.
  • the cultured cell may be a cultured cell transfected with the vector or a progeny of the cell, wherein the cell expresses the immunogenic polypeptide.
  • Suitable cell lines are known to those of skill in the art and are commercially available, for example, through the American Type Culture Collection (ATCC).
  • ATCC American Type Culture Collection
  • the transfected cells can be used in a method of producing an immunogenic polypeptide.
  • the method comprises culturing a cell comprising the vector under conditions that allow expression of the immunogenic polypeptide, optionally under the control of an expression sequence.
  • the immunogenic polypeptide can be isolated from the cell or the culture medium using standard protein purification methods.
  • polypeptides and nucleic acids described herein may be combined with one or more pharmaceutically acceptable carriers prior to administration to a host.
  • a pharmaceutically acceptable carrier is a material that is not biologically or otherwise undesirable, i.e., the material may be administered to a subject, without causing any undesirable biological effects or interacting in a deleterious manner with any of the other components of the pharmaceutical composition in which it is contained.
  • the carrier would naturally be selected to minimize any degradation of the active ingredient and to minimize any adverse side effects in the subject, as would be well known to one of skill in the art.
  • Suitable pharmaceutical carriers and their formulations are described in, for example, Remington's: The Science and Practice of Pharmacy, 21 st Edition , David B. Troy, ed., Lippicott Williams & Wilkins (2005).
  • an appropriate amount of a pharmaceutically-acceptable salt is used in the formulation to render the formulation isotonic.
  • the pharmaceutically-acceptable carriers include, but are not limited to, sterile water, saline, buffered solutions like Ringer's solution, and dextrose solution. The pH of the solution is generally from about 5 to about 8 or from about 7 to about 7.5.
  • Other carriers include sustained-release preparations such as semipermeable matrices of solid hydrophobic polymers containing polypeptides or fragments thereof.
  • Matrices may be in the form of shaped articles, e.g., films, liposomes or microparticles. It will be apparent to those persons skilled in the art that certain carriers may be more preferable depending upon, for instance, the route of administration and concentration of composition being administered. Carriers are those suitable for administration of polypeptides and/or fragments thereof to humans or other subjects.
  • compositions may also include carriers, thickeners, diluents, buffers, preservatives, surface active agents, adjuvants, immunostimulants, in addition to the immunogenic polypeptide.
  • Pharmaceutical compositions may also include one or more active ingredients such as antimicrobial agents, antiinflammatory agents and anesthetics.
  • Adjuvants may also be included to stimulate or enhance the immune response.
  • suitable classes of adjuvants include those of the gel-type (i.e., aluminum hydroxide/phosphate (“alum adjuvants”), calcium phosphate, microbial origin (muramyl dipeptide (MDP)), bacterial exotoxins (cholera toxin (CT), native cholera toxin subunit B (CTB), E.
  • LT coli labile toxin
  • PT pertussis toxin
  • CpG oligonucleotides BCG sequences, tetanus toxoid, monophosphoryl lipid A (MPL) of, for example, E.
  • coli Salmonella minnesota, Salmonella typhimurium , or Shigella exseri
  • particulate adjuvants biodegradable, polymer microspheres
  • ISCOMs immunostimulatory complexes
  • FIA oil-emulsion and surfactant-based adjuvants
  • MF59, SAF microfluidized emulsions
  • QS-21 saponins
  • synthetic (muramyl peptide derivatives murabutide, threony-MDP
  • nonionic block copolymers L121
  • PCCP polyphosphazene
  • synthetic polynucleotides poly A:U, poly I:C
  • thalidomide derivatives CC-4407/ACTIMID
  • RH3-ligand or polylactide glycolide (PLGA) microspheres, among others.
  • Fragments, homologs, derivatives, and fusions to any of these toxins are also suitable, provided that they retain adjuvant activity.
  • Suitable mutants or variants of adjuvants are described, e.g., in WO 95/17211 (Arg-7-Lys CT mutant), WO 96/6627 (Arg-192-Gly LT mutant), and WO 95/34323 (Arg-9-Lys and Glu-129-Gly PT mutant).
  • Additional LT mutants that may used include, for example, Ser-63-Lys, Ala-69-Gly,Glu-110-Asp, and Glu-112-Asp mutants.
  • Metallic salt adjuvants such as alum adjuvants are well-known in the art as providing a safe excipient with adjuvant activity.
  • the mechanism of action of these adjuvants are thought to include the formation of an antigen depot such that antigen may stay at the site of injection for up to 3 weeks after administration, and also the formation of antigen/metallic salt complexes which are more easily taken up by antigen presenting cells.
  • other metallic salts have been used to adsorb antigens, including salts of zinc, calcium, cerium, chromium, iron, and berilium.
  • the hydroxide and phosphate salts of aluminium are the most common.
  • Formulations or compositions containing aluminium salts, antigen, and an additional immunostimulant are known in the art.
  • An example of an immunostimulant is 3-de-O-acylated monophosphoryl lipid A (3D-MPL).
  • mPLY polypeptides and/or fragments thereof may be covalently coupled to bacterial polysaccharides to form polysaccharide conjugates.
  • Such conjugates may be useful, for example, as immunogens for eliciting a T cell dependent immunogenic response directed against the bacterial polysaccharide conjugated to the mPLY and/or fragments thereof.
  • cytokines may also be suitable co-stimulatory components for use with the mPLY polypeptides and/or fragments thereof, either as polypeptides or as encoded by nucleic acids (Parmiani, et al. Immunol Lett 2000 Sep. 15; 74(1): 41-3; Berzofsky, et al. Nature Immunol. 1: 209-219).
  • Suitable cytokines include, for example, interleukin-2 (IL-2) (Rosenberg, et al. Nature Med. 4: 321-327 (1998)), IL-4, IL-7, IL-12 (reviewed by Pardoll, 1992; Harries, et al. J. Gene Med.
  • cytokines may also be suitable for use, as is known in the art.
  • antibody or “antibodies” includes whole or fragmented antibodies in unpurified or partially purified form (i.e., hybridoma supernatant, ascites, polyclonal antisera) or in purified form.
  • a “purified” antibody is one that is separated from at least about 50% of the proteins with which it is initially found (i.e., as part of a hybridoma supernatant or ascites preparation).
  • a purified antibody is separated from at least about 60%, 75%, 90%, or 95% of the proteins with which it is initially found.
  • Suitable derivatives may include fragments (i.e., Fab, Fab 2 or single chain antibodies (Fv for example)), as are known in the art.
  • the antibodies may be of any suitable origin or form including, for example, murine (i.e., produced by murine hybridoma cells), or expressed as humanized antibodies, chimeric antibodies, human antibodies, and the like.
  • the antibodies may be contained within hybridoma supernatant or ascites and utilized either directly as such or following concentration using standard techniques.
  • the antibodies may be further purified using, for example, salt fractionation and ion exchange chromatography, or affinity chromatography using Protein A, Protein G, Protein A/G, and/or Protein L ligands covalently coupled to a solid support such as agarose beads, or combinations of these techniques.
  • the antibodies may be stored in any suitable format, including as a frozen preparation (i.e., about ⁇ 20° C. or ⁇ 70° C.), in lyophilized form, or under normal refrigeration conditions (i.e., about 4° C.).
  • TBS Tris-buffered saline
  • PBS phosphate buffered saline
  • Antibodies and their derivatives may be incorporated into compositions described herein for use in vitro or in vivo. Other methods for making and using antibodies available to one of skill in the art may also be suitable for use.
  • kits for detecting the presence of streptococcus infection in a patient by detecting antibodies or nucleic acid in a biological sample of the patient may form part of a kit for detecting or diagnosing anti-streptococcal antibodies in a biological sample.
  • the antigens may be provided in a suitable container such as a vial in which the contents are protected from the external environment.
  • kits for detecting an anti-streptococcal antibody in a sample may comprise one or more mutant PLY polypeptides and/or fragments thereof along with one or more detection reagents for determining binding of one or more antibodies in a sample to the antigen is provided.
  • the kit preferably includes: (i) one or more isolated and purified mPLY polypeptides and/or fragments thereof; and, (ii) a system for detecting the formation of an antigen-antibody complex, optionally packaged with instructions for use.
  • the antigen may be free in solution or may be immobilized on a solid support, such as a magnetic bead, tube, microplate well, or chip.
  • a solid matrix comprising an isolated and purified mPLY polypeptides and/or fragments thereof or a fusion protein or protein aggregate adsorbed thereto.
  • the kit may further comprise an antibody-binding molecule as a detection reagent.
  • the antibody-binding molecule may be a capture or detection reagent and may be free in solution or may be immobilized on a solid support, such as a magnetic bead, tube, microplate well, or chip.
  • the antibody-binding molecule or polypeptide may be labeled with a detectable label, for example a fluorescent or chromogenic label or a binding moiety such as biotin. Suitable labels are described in more detail above.
  • the kit may further comprise detection reagents such as a substrate, for example a chromogenic, fluorescent or chemiluminescent substrate, which reacts with the label, or with molecules, such as enzyme conjugates, which bind to the label, to produce a signal, and/or reagents for immunoprecipitation (i.e., protein A or protein G reagents).
  • the detection reagents may further comprise buffer solutions, wash solutions, and other useful reagents.
  • the kit may also comprise one or both of an apparatus for handling and/or storing the sample obtained from the individual and an apparatus for obtaining the sample from the individual (i.e., a needle, lancet, and collection tube or vessel).
  • the kit may also include instructions for use of the antigen, e.g.
  • the assay is to be combined with another type of assay such as PCR, the required reagents for such an assay (i.e., primers, buffers and the like) along with, optionally, instructions for the use thereof, may also be included.
  • a subject or a host is meant to be an individual.
  • the subject can include domesticated animals, such as cats and dogs, livestock (e.g., cattle, horses, pigs, sheep, and goats), laboratory animals (e.g., mice, rabbits, rats, guinea pigs) and birds.
  • livestock e.g., cattle, horses, pigs, sheep, and goats
  • laboratory animals e.g., mice, rabbits, rats, guinea pigs
  • the subject is a mammal such as a primate or a human.
  • composition can comprise a combination means that the composition may comprise a combination of different molecules or may not include a combination such that the description includes both the combination and the absence of the combination (i.e., individual members of the combination).
  • Ranges may be expressed herein as from about one particular value, and/or to about another particular value. When such a range is expressed, another aspect includes from the one particular value and/or to the other particular value. Similarly, when values are expressed as approximations, by use of the antecedent about or approximately, it will be understood that the particular value forms another aspect. It will be further understood that the endpoints of each of the ranges are significant both in relation to the other endpoint, and independently of the other endpoint. Ranges (e.g., 90-100%) are meant to include the range per se as well as each independent value within the range as if each value was individually listed.
  • prevent, preventing, and prevention are used herein in connection with a given treatment for a given condition (e.g., preventing infection by Streptococcus sp.), it is meant to convey that the treated patient either does not develop a clinically observable level of the condition at all, or develops it more slowly and/or to a lesser degree than he/she would have absent the treatment.
  • a treatment will be said to have prevented the condition if it is given during exposure of a patient to a stimulus that would have been expected to produce a given manifestation of the condition, and results in the patients experiencing fewer and/or milder symptoms of the condition than otherwise expected.
  • a treatment can “prevent” infection by resulting in the patients displaying only mild overt symptoms of the infection; it does not imply that there must have been no penetration of any cell by the infecting microorganism.
  • reduce, reducing, and reduction as used herein in connection with the risk of infection with a given treatment typically refers to a subject developing an infection more slowly or to a lesser degree as compared to a control or basal level of developing an infection in the absence of a treatment (e.g., administration or vaccination using mPLY).
  • a reduction in the risk of infection may result in the patients displaying only mild overt symptoms of the infection or delayed symptoms of infection; it does not imply that there must have been no penetration of any cell by the infecting microorganism.
  • the ply gene encoding wtPLY was cloned by PCR using primers Spn001 (CATGCCATGGCAAATAAAGCAGTAAATGAC; SEQ ID NO. 45) and Spn002 (CAGCCGCTCGAGCTAGTCATTTTCTACCTTATCCTC; SEQ ID NO. 46) from S. pneumoniae strain R36A (SEQ ID NO. 1, GenBank Accession No. M17717).
  • the PCR product was digested with restriction enzymes NcoI and XhoI and cloned into plasmid pTrcK.
  • Plasmid pTrcK is a kanamycin-resistant derivative of plasmid pTrcHis2 (Invitrogen).
  • the plasmid thus generated (pBM46) was used for expression of wtPLY (producing a non-tagged PLY polypeptide) under control of the trc promoter.
  • the DNA sequence of the amplicon was identical to the published ply sequence shown in SEQ ID NO. 1), encoding the deduced amino acid sequence shown in SEQ ID NO. 2.
  • the wtPLY-encoding nucleic acid was then transferred into pET-28a (EMD Biosciences Cat. No. 69864-3) (NcoI-XhoI), resulting in plasmid pJMS102 (providing non-tagged wtPLY expression from the T7 promoter).
  • Plasmid pJMS112 from Example 1 was used as the template for site-directed mutagenesis to modify the expressed PLY polypeptide.
  • Site-directed mutagenesis was performed using the QuickChange Multi Site-Directed Mutagenesis kit as per the manufacturer's instructions (Agilent Technologies, Stratagene Products Division, La Jolla, Calif.). The following modifications were introduced into the PLY encoding nucleotide sequence:
  • Thr65 was changed from ACC to TGC, encoding cysteine (C) in place of threonine (T) (T65C);
  • Plasmid pJAY7 was digested with NdeI and XhoI to isolate the 1420 by ply gene therefrom. This 1420 bp fragment was gel purified and then ligated into NdeI-XhoI cut and dephosphorylated pET-30a (EMD Biosciences Cat. No. 69909-3). The resulting plasmid was named pJMS140, and provided non-tagged PlyD1 expression from the T7 promoter. The sequence of the mutated ply gene encoding PlyD1 was confirmed using the primers shown in Table 3 below.
  • PlyD1 SEQ ID NO. 44; T65C, G293C, C428A was expressed as a recombinant protein ( ⁇ 55 kDa) in pET30A plasmid using an E. coli expression system.
  • the E. coli expressed recombinant PlyD1 protein was grown in a 20-L fermentor. The cells were homogenized at high pressure to release the soluble PlyD1 and diafiltered in Tris buffered saline to produce a crude extract of PlyD1.
  • the crude PlyD1 extract was 0.2 um filtered prior to use. The filtered, crude extract was run through the strong anion exchange Giga Cap Q-650 M column (Tosoh Biosciences).
  • Unbound contaminant proteins were removed in the flow-through and chased with equilibration buffer (20 mM Tris-HCl, pH 8.5) An intermediate wash step was performed using equilibration buffer containing 100 mM NaCl. Bound PlyD1 was eluted with 20 mM Tris-HCl, pH 8.5 containing 250 mM NaCl, PlyD1 material recovered from this column chromatography step was then conditioned with 5 M NaCl solution to bring the conductivity up to ⁇ 80 mS/cm.
  • the conditioned PlyD1 material was then subjected to a hydrophobic column chromatography using Phenyl Sepharose FF (GE Health Care). PlyD1 was purified using binding and elution mode. Unbound proteins were removed by washing the column with 1M NaCl equilibration buffer and bound PlyD1 eluted with the 20 mM Tris-HCl buffer, pH 8.0
  • the phenyl sepharose-purified PlyD1 material was then diluted 4-fold with 5 mM sodium phosphate (pH 6.2) to decrease the conductivity and pH of the binding solution. This material was then further purified using Ceramic Hydroxyapatite type I 40 um resin (BioRad).
  • the equilibration buffer is 5 mM Sodium Phosphate, pH 6.2.
  • PlyD1 was purified a second time using a binding and elution mode on this mixed mode column chromatography resin. Contaminants were removed using 5 mM sodium phosphate, pH 7.0, 750 mM NaCl and bound PlyD1 eluted with 10 mM sodium phosphate, pH 7.0, containing 1 M NaCl.
  • purified PlyD1 was diafiltered in 10 mM Tris-HCl, pH 7.4, 150 mM NaCl buffer. Tween-80 was added to a final concentration of 0.05% to prevent PlyD1 precipitation.
  • the purified PlyD1 bulk material was 0.2 ⁇ M filtered and subsequently formulated at different concentrations. A typical purification process yields 600-850 mg/L of purified PlyD1 protein with a purity of >98% based on SDS-PAGE analysis ( FIG. 4 ).
  • Cytolytic activity of PLY polypeptides is customarily evaluated by an in vitro hemolysis assay.
  • test proteins are serially diluted across a plastic microtiter plate in 2-fold serial dilutions with a highest concentration of 0.5 mg/mL of test protein.
  • BSA is included to prevent adsorptive losses on the plastic microtiter plate.
  • Sheep red blood cells are added to all wells, and the plastic microtiter plates are incubated for 30 min. Lysed cells release hemoglobin.
  • 100% lysis measurement is obtained by the addition of 1% Trition X-100.
  • the sheep red blood cells are incubated with PBS alone.
  • the microtiter plate is centrifuged to separate the intact cells from the lysed cells.
  • the supernatant containing the lysed cells are transferred to a fresh plate and subject to an A 540 hemoglobin release assay.
  • the specific activity is determined as the inverse of protein concentration (mg/mL) at which 50% hemolysis has occurred relative to the positive control.
  • a representative hemolytic assay system is shown below:
  • PlyD1 was expressed in E. coli and purified as described above. The purified proteins were dialyzed against PBS and used for the immunization of rabbits. Two rabbits were immunized intra-muscularly at 20 ⁇ g dose with Freund's adjuvant. The animals received two additional immunizations with incomplete Freund's adjuvant two and four weeks after the initial injection. Serum was collected two and four weeks after the last injection. The generation of antibodies in the rabbit was first tested by western blot to ensure that anti-PlyD1 antibodies react with wtPLY. Rabbit sera may also be tested by ELISA and/or IGEN competition assay using standard procedures to detect the presence of antibodies specific for known neutralizing epitopes.
  • the assay to determine the ability of anti-PlyD1 antibodies to inhibit wtPLY hemolytic activity involves the following steps. First, antisera are produced essentially as described herein then serially diluted two-fold across a microtiter plate. A constant amount of native wtPLY is added to all wells. Sheep red blood cells are added to all wells and the assay proceeds as per the in vitro hemolysis assay described above. After final plate processing, the data are graphed to determine the 50% inhibition titer. A representative haemolysis-inhibition assay system is shown below:
  • PLY mutants including Ply-G293A (G293 substituted by alanine), Ply-G293T (G293 substituted by threonine), Ply-G293V (G293 substituted by valine), and Ply-G293C (G293 substituted by cysteine) also showed lower hemolytic activity (including no detectable activity) than wtPLY (“Ply” in FIG. 4 ). Additional independent lots of PlyD1 were also tested. Hemolytic activity was not detected in any of the PlyD1 lots.
  • PlyD1 was administered in varying doses, with or without aluminum hydroxide (AlOOH) adjuvant. PlyD1 was found to induce high anti-PLY IgG titers and hemolysis inhibition (HI) titers at all doses tested.
  • AlOOH aluminum hydroxide
  • PlyD1 was administered with AlOOH adjuvant following two-weeks storage at ⁇ 70° C., 2-8° C., or 45° C.
  • PlyD1 was administered in varying doses, with or without AlOOH adjuvant.
  • Mice were immunized three times, three weeks apart, and blood samples were taken three weeks after each immunization.
  • Antibody titers from the second and final bleeds were measured using PLY-specific ELISA.
  • ELISA titers showed a dose response for anti-PLY titers following the second and third bleed. Immunization with adjuvanted PlyD1 resulted in significantly higher anti-PLY titers compared to those induced by unadjuvanted PlyD1.
  • ELISA results from the third bleed are shown in Table 5.
  • the lower limit of detection of the assay is 4; titers lower than 4 are listed as 2 for statistical purposes.
  • PlyD1 generated high anti-PLY IgG titers when PlyD1, stored at either ⁇ 70° C. or 2-8° C. was used for immunization, but not PlyD1 stored at 45° C.
  • PlyD1 was able to generate higher titers of anti-PLY-specific antibodies in comparison to PlyD1 administered without adjuvant.
  • wtPLY is known to activate macrophages to generate cytokines (Malley, et al. PNAS USA, 100(4):1966-71 (2003)).
  • This assay determines macrophage activation by PlyD1 by measuring PlyD1-induced cytokine production in vitro. J774 (mouse) and MM6 (human) macrophage-like cells were incubated overnight with PdB, PlyD1 or PLY treated (+/+) or not treated ( ⁇ / ⁇ ) with proteinase K and heat. Heat was used in order to distinguish any false positive results due to lipopolysaccharide (LPS) contamination in the treated group.
  • LPS lipopolysaccharide
  • Cytokine production (IL-6, IL-10, TNF- ⁇ , IL-1 ⁇ ) was measured by ELISA after overnight incubation. No induction of cytokines (IL-1 ⁇ , IL-6 and IL-10) was detected following co-culture of mouse or human macrophage cell lines (J774A.1 or MM6, respectively) with PlyD1 either untreated or treated with proteinase K/heat. In comparison, untreated wtPLY was able to induce low amounts of IL-1 ⁇ and IL-6 cytokine release in MM6 cells (PLY ⁇ / ⁇ ).
  • mice The ability of PlyD1 to induce anti-PLY antibodies and induce a protective immune response in mice was assessed.
  • PdB was administered at 10 ⁇ g/dose and PlyD1 at 1 ⁇ g, 2.5 ⁇ g, and 10 ⁇ g/dose.
  • the injection volume was 200 ⁇ L per dose.
  • TBS placebo-containing aluminum adjuvant was injected into negative control groups. Animals were immunized SC at 0, three and six weeks following initiation of the study.
  • mice were challenged by intranasal (IN) injection with 10 7 colony forming units (cfu) of S. pneumoniae serotype 6B in phosphate-buffered saline (PBS) suspension (50 ⁇ L challenge volume per mouse). Following the challenge, mice were monitored for mortality daily. Fourteen days post-challenge, all surviving mice were euthanized. The Fisher Exact Test was used to determine if there was a significant difference between the immunized group(s) and the placebo control. In addition, sample bleeds were taken from all animals following the second boost (day 42) and prior to challenge, following three immunizations (day 63). Sera were analyzed for total pneumolysin-specific IgG response by means of an antibody ELISA and for pneumolysin neutralizing capacity in an inhibition of hemolysis assay.
  • PBS phosphate-buffered saline
  • none of the groups immunized with PlyD1 showed significant survival or delay to death compared to the placebo control group (p>0.05) (Table 7).
  • PlyD1 was also tested using a focal pneumonia mouse model. Briefly, groups of 10 CBA/N mice were immunized SC with purified recombinant PlyD1 proteins at variable doses formulated in TBS-containing aluminum adjuvant (300 ⁇ g/dose). The injection volume was 200 ⁇ L per dose. Phosphate-buffered saline placebo-containing aluminum adjuvant was injected into negative control groups. Animals were immunized SC three times at 0, three and six weeks following initiation of the study. Three weeks after the last immunization, animals were challenged intranasally (IN) with 3 ⁇ 7 ⁇ 10 6 cfu of S. pneumoniae strain EF3030 (serotype 19F; 40 ⁇ L challenge volume per mouse).
  • IN intranasally
  • mice were sacrificed 5 days post-challenge and lung tissue harvested and plated for cfu recovery.
  • the Mann-Whitney Test was used to determine if there was a significant difference between immunized group(s) and the placebo control group. In studies using this model, sera analysis for either IgG titer or neutralizing capacity of the sera was not performed. All groups that were immunized with PlyD1 did not have significantly lower bacterial lung burden when compared to the PBS-immunized group and thus, were considered as not protected (data not shown).
  • PlyD1 was also evaluated in-house using an intranasal challenge mouse model.
  • PlyD1 was formulated in TBS-containing aluminum adjuvant (65 ⁇ g/dose). The injection volume was 50 ⁇ L per dose.
  • PBS placebo-containing aluminum adjuvant was injected into negative control groups. Animals were immunized IM at 0, 3, and 6 weeks following initiation of the study. At nine weeks, animals were challenged IN with a lethal dose (5 ⁇ 10 5 cfu) of S.
  • pneumoniae strain 14453 (serotype 6B) in PBS suspension (40 ⁇ L challenge volume per mouse). Following the challenge, mice were monitored daily for mortality. All surviving mice were euthanized 11 days post-challenge. The Fisher Exact Test was used to determine if there was a significant difference between the immunized group(s) and the placebo control. In addition, sample bleeds were taken from all animals 4 days prior to the first injection (pre-immunization at 0 weeks) and following each immunization. Sera were analyzed for total PlyD1-specific IgG responses by means of an antibody ELISA and for pneumolysin neutralizing capacity in an inhibition of hemolysis assay.
  • PlyD1 was administered at 0.25 ⁇ g, 0.5 ⁇ g, 1 ⁇ g, 5 ⁇ g, 10 ⁇ g and 25 ⁇ g/dose or an adjuvant alone (placebo). Mice were immunized three times, three weeks apart and blood samples were taken three weeks following each immunization. Antibody titers from the second bleed and final bleed (three weeks following last immunization) were measured using PLY-specific ELISA. The ability of the first, second and final bleed sera to inhibit PLY-mediated hemolysis was assessed. Mice were challenged with S. pneumoniae strain 14453 (serotype 6B) at 5 ⁇ 10 5 cfu/dose three weeks following the last immunization and survival was monitored for ten days.
  • S. pneumoniae strain 14453 serotype 6B
  • mice immunized with PlyD1 was significantly better than placebo group at doses of 5, 2.5 and 0.25 ⁇ g/dose, indicating that protection is not dose-dependent. The best protection was observed at 2.5 ⁇ g/dose, in which 60% of the mice survived compared to none in the placebo group. The results are summarized in Table 9.
  • mice immunized using this intranasal challenge model were also assessed for inhibition of PLY-mediated hemolysis of sheep red blood cells.
  • Sera from all bleeds were pooled according to their treatment groups and tested using the HI assay following the all bleeds. While the HI titers appeared to be slightly higher after the third immunization (bleed 3) in mice immunized with 2.5 and 5 ⁇ g/dose, this difference is considered to be within the assay variation and therefore is not significant. Furthermore, there did not appear to be a dose response in the HI titers generated with increasing amounts of PlyD1. The results are shown in Table 10.
  • Antibody titers from the second bleed and third bleed were measured using quantitative anti-PLY ELISA. All PlyD1 immunized mice were able to mount an antibody response to PLY following bleed 2 and bleed 3. There were no significant differences ( ⁇ 2 fold differences) observed between groups immunized with increasing doses of PlyD1, suggesting that all doses tested were still generating saturating amounts of anti-PLY antibodies.
  • ELISA results from the third bleed are shown in Table 11.
  • mice were also immunized with PlyD1 with or without adjuvant, wtPLY at a dose of 5 or 10 ⁇ g, and then challenged with a 5 ⁇ g dose of wtPLY. The lungs were then harvested for H&E stain to observe tissue damage caused by wtPLY. Compared to controls (mice immunized with Tris-saline, 15% glycerol), wtPLY typically causes perivascular edema, thickened, disrupted alveolar walls, diminished alveolar space, and fluid and blood infiltration of the alveolar spaces. As shown in FIG.
  • mice immunized with PlyD1, PdB, or wtPLY demonstrated significant protection from lung damage after intranasal challenge with wtPLY.
  • the ability of antisera from these immunized mice to inhibit PLY-mediated hemolysis of sheep red blood cells was tested using the HI assay following the first, second and third bleed.
  • Neutralizing antibody titers were increased following bleed 3 compared to bleed 2.
  • PlyD1 immunization can generate antibodies that are directly involved in the neutralization of PLY toxicity in vivo.
  • mice were immunized (I.M) with adjuvant alone (AlOOH) or with monovalent Ply-D1, 3 times with 3 weeks interval between each immunization. 3 weeks post final immunization, mice were challenged with GT-14453.BM2 (serotype 6B) at 5 ⁇ 10 5 CFU/mouse and observed for survival/health for 2 weeks.
  • PlyD1 was formulated as 0.25 ⁇ g, 0.5 ⁇ g, 1.0 ⁇ g, 2.5 ⁇ g, 5.0 ⁇ g, 10 ⁇ g and 25 ⁇ g/ml. Data was analyzed by plotting survival/health status of mice and statistically compared using Fisher-one sided test for statistical analysis. As shown in FIG. 5 , immunization with recombinant PlyD1 led to protection against lethal challenge in mice.
  • mice immunized with PlyD1 had significantly lower lung damage when compared to placebo-immunized and PLY challenged mice.
  • the production process for a PlyD1 immunogenic composition and/or vaccine involves: 1) growing recombinant E. coli cells that express the PlyD1 protein in a fermenter, using pH-Stat fed batch fermentation; 2) recovering the PlyD1 protein by homogenizing cells followed by 0.2 ⁇ m clarification filtration, and concentration by ultrafiltration; 3) purifying the PlyD1 protein using ion-exchange chromatography and hydrophobic interaction chromatography; and, 4) formulating the purified PlyD1 protein with aluminum adjuvant, however other adjuvants known in the art an also be used. These procedures are described herein or well-known to those of skill in the art of vaccine formulation.
  • PlyD1 may be formulated with an aluminum hydroxide adjuvant in sterile Tris-buffered saline (TBS) without preservative, and prepared as a sterile, white opaque liquid suspension in single-dose vials (containing 0.28 mg of elemental aluminum/dose).
  • TBS Tris-buffered saline
  • Other adjuvants known in the art can also be used to formulate a vaccine.
  • Each 0.5 mL dose of PlyD1 immunogenic composition or vaccine typically contains the following components: recombinant PlyD1 (10 ⁇ g (low dose), 25 ⁇ g (middle dose), 50 ⁇ g (high dose)), and Tris-buffered saline (TBS; 10 mM Tris-HCl pH 7.4, 150 mM sodium chloride), aluminum hydroxide adjuvant (0.28 mg elemental aluminum/dose), and sodium phosphate (2 mM) to optimize binding and stability of adsorbed antigens.
  • TBS Tris-buffered saline
  • the glass vials are filled with 0.72 ⁇ 0.05 mL to give a withdrawable volume of 0.50 mL.
  • An injection (intramuscular, IM) of 0.5 mL from a low dose, middle dose and high dose will give a dosage of either 10 ⁇ g, 25 ⁇ g or 50 ⁇ g of PlyD1, respectively.
  • Each 0.5 mL dose is adjuvanted with 0.28 mg ⁇ 0.10 mg elemental aluminum.
  • the placebo to be used is TBS.
  • the immunogenic composition or vaccine is typically supplied in 3 mL glass vials that have a 13 mm gray butyl serum stopper that is latex-free and a 13 mm one-piece aluminum seal.
  • the composition is typically a white cloudy suspension and the placebo is a clear solution and must be stored at about 2° C. to 8° C. (e.g., not frozen).
  • the aluminum adjuvant in the product typically settles over time and is re-suspended before use.
  • the vaccine vial In preparation for use, the vaccine vial should be inverted about 5 to 10 times until the contents are uniform in appearance.
  • a syringe should be filled immediately after suspension/mixing and the vaccine injected promptly.
  • Routes of administrations may be as described herein, for example, preferably via subcutaneous (SC), intradermal (ID), intramuscular (IM), or oral routes.
  • SEQUENCE LISTING (pBM46 nucleotide sequence wtPLY insert S. pneumoniae R36A) SEQ ID NO.: 1 ATGGCAAATAAAGCAGTAAATGACTTTATACTAGCTATGAATTACGATAAAAAGAAACTCTTGACCCATCA GGGAGAAAGTATTGAAAATCGTTTCATCAAAGAGGGTAATCAGCTACCCGATGAGTTTGTTGTTATCGAAA GAAAGAAGCGGAGCTTGTCGACAAATACAAGTGATATTTCTGTAACAGCTACCAACGACAGTCGCCTCTAT CCTGGAGCACTTCTCGTAGTGGATGAGACCTTGTTAGAGAATAATCCCACTCTTCTTGCGGTTGATCGTGC TCCGATGACTTATAGTATTGATTTGCCTGGTTTGGCAAGTAGCGATAGCTTTCTCCAAGTGGAAGACCCCA GCAATTCAAGTGTTCGCGGAGCGGTAAACGATTTGTTGGCTAAGTGGCATCAAGATTATGGTCAGGTCAAT AATGTCCCAGCTAGAATG

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Immunology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Epidemiology (AREA)
  • Mycology (AREA)
  • Microbiology (AREA)
  • Pulmonology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Oncology (AREA)
  • Communicable Diseases (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)
US13/140,894 2008-12-24 2009-12-22 Modified Streptococcus pneumoniae pneumolysin (PLY) polypeptides Expired - Fee Related US8758766B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US13/140,894 US8758766B2 (en) 2008-12-24 2009-12-22 Modified Streptococcus pneumoniae pneumolysin (PLY) polypeptides

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US14080408P 2008-12-24 2008-12-24
PCT/CA2009/001843 WO2010071986A1 (en) 2008-12-24 2009-12-22 Modified steptococcus pneumonia pneumolysin (ply) polypeptides
US13/140,894 US8758766B2 (en) 2008-12-24 2009-12-22 Modified Streptococcus pneumoniae pneumolysin (PLY) polypeptides

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/CA2009/001843 A-371-Of-International WO2010071986A1 (en) 2008-12-24 2009-12-22 Modified steptococcus pneumonia pneumolysin (ply) polypeptides

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/269,501 Continuation US8951531B2 (en) 2008-12-24 2014-05-05 Modified Streptococcus pneumonia pneumolysin (PLY) polypeptides

Publications (2)

Publication Number Publication Date
US20110287046A1 US20110287046A1 (en) 2011-11-24
US8758766B2 true US8758766B2 (en) 2014-06-24

Family

ID=42286822

Family Applications (3)

Application Number Title Priority Date Filing Date
US13/140,894 Expired - Fee Related US8758766B2 (en) 2008-12-24 2009-12-22 Modified Streptococcus pneumoniae pneumolysin (PLY) polypeptides
US14/269,501 Expired - Fee Related US8951531B2 (en) 2008-12-24 2014-05-05 Modified Streptococcus pneumonia pneumolysin (PLY) polypeptides
US14/575,541 Expired - Fee Related US9546199B2 (en) 2008-12-24 2014-12-18 Modified Streptococcus pneumoniae pneumolysin (PLY) polypeptides

Family Applications After (2)

Application Number Title Priority Date Filing Date
US14/269,501 Expired - Fee Related US8951531B2 (en) 2008-12-24 2014-05-05 Modified Streptococcus pneumonia pneumolysin (PLY) polypeptides
US14/575,541 Expired - Fee Related US9546199B2 (en) 2008-12-24 2014-12-18 Modified Streptococcus pneumoniae pneumolysin (PLY) polypeptides

Country Status (11)

Country Link
US (3) US8758766B2 (he)
EP (1) EP2376526A4 (he)
KR (1) KR101741426B1 (he)
CN (2) CN106008679A (he)
AU (1) AU2009329782B2 (he)
BR (1) BRPI0923649A2 (he)
CA (1) CA2748149C (he)
IL (2) IL213734A (he)
MX (1) MX2011006922A (he)
WO (1) WO2010071986A1 (he)
ZA (1) ZA201104627B (he)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10562941B2 (en) 2014-11-21 2020-02-18 The Board Of Regents Of The University Of Oklahoma Pneumolysin mutants and methods of use thereof
US10973908B1 (en) 2020-05-14 2021-04-13 David Gordon Bermudes Expression of SARS-CoV-2 spike protein receptor binding domain in attenuated salmonella as a vaccine

Families Citing this family (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20130183350A1 (en) * 2009-12-22 2013-07-18 Kevin Harper Immunogenic compositions
CN102939105B (zh) 2009-12-22 2016-11-16 赛诺菲巴斯德有限公司 免疫原性组合物和相关的方法
GB201003924D0 (en) 2010-03-09 2010-04-21 Glaxosmithkline Biolog Sa Immunogenic composition
WO2012075428A1 (en) * 2010-12-03 2012-06-07 Sanofi Pasteur Limited Composition for immunization against streptococcus pneumoniae
WO2012155053A1 (en) * 2011-05-11 2012-11-15 Children's Medical Center Corporation Modified biotin-binding protein, fusion proteins thereof and applications
JP2014515035A (ja) 2011-05-17 2014-06-26 グラクソスミスクライン バイオロジカルズ ソシエテ アノニム ストレプトコッカス・ニューモニエに対するワクチン
WO2013063283A1 (en) 2011-10-25 2013-05-02 Corning Incorporated Delamination resistant pharmaceuticals glass containers containing active pharmaceutical ingredients
GB201218660D0 (en) 2012-10-17 2012-11-28 Glaxosmithkline Biolog Sa Immunogenic composition
MX365842B (es) * 2012-10-17 2019-06-17 Glaxosmithkline Biologicals Sa Composicion inmunogenica que comprende 1 o mas conjugados de sacaridos capsulares de streptococcus pneumoniae y un componente proteico que comprende proteinas e y/o pila de haemophilus influenzae.
WO2014124228A1 (en) 2013-02-07 2014-08-14 Children's Medical Center Corporation Protein antigens that provide protection against pneumococcal colonization and/or disease
US9849066B2 (en) 2013-04-24 2017-12-26 Corning Incorporated Delamination resistant pharmaceutical glass containers containing active pharmaceutical ingredients
US9717649B2 (en) 2013-04-24 2017-08-01 Corning Incorporated Delamination resistant pharmaceutical glass containers containing active pharmaceutical ingredients
US9603775B2 (en) 2013-04-24 2017-03-28 Corning Incorporated Delamination resistant pharmaceutical glass containers containing active pharmaceutical ingredients
US9717648B2 (en) 2013-04-24 2017-08-01 Corning Incorporated Delamination resistant pharmaceutical glass containers containing active pharmaceutical ingredients
US9713572B2 (en) 2013-04-24 2017-07-25 Corning Incorporated Delamination resistant pharmaceutical glass containers containing active pharmaceutical ingredients
US9839579B2 (en) 2013-04-24 2017-12-12 Corning Incorporated Delamination resistant pharmaceutical glass containers containing active pharmaceutical ingredients
US9707155B2 (en) 2013-04-24 2017-07-18 Corning Incorporated Delamination resistant pharmaceutical glass containers containing active pharmaceutical ingredients
US9700485B2 (en) 2013-04-24 2017-07-11 Corning Incorporated Delamination resistant pharmaceutical glass containers containing active pharmaceutical ingredients
US9707153B2 (en) * 2013-04-24 2017-07-18 Corning Incorporated Delamination resistant pharmaceutical glass containers containing active pharmaceutical ingredients
US9700486B2 (en) 2013-04-24 2017-07-11 Corning Incorporated Delamination resistant pharmaceutical glass containers containing active pharmaceutical ingredients
US9707154B2 (en) 2013-04-24 2017-07-18 Corning Incorporated Delamination resistant pharmaceutical glass containers containing active pharmaceutical ingredients
GB201610599D0 (en) * 2016-06-17 2016-08-03 Glaxosmithkline Biologicals Sa Immunogenic Composition
CA3061709A1 (en) 2017-03-28 2018-10-04 The Children's Medical Center Corporation A multiple antigen presenting system (maps)-based staphylococcus aureus vaccine, immunogenic composition, and uses thereof
EP3641828B1 (en) 2017-06-23 2023-11-22 Affinivax, Inc. Immunogenic compositions
KR102076417B1 (ko) * 2018-08-01 2020-02-11 윤현규 측방유동분석법을 이용한 세균성 폐렴균의 검출 방법 및 키트
BR112021004193A2 (pt) 2018-09-12 2021-05-25 Affinivax, Inc. vacinas pneumocócicas multivalentes
EP3747903A1 (en) * 2019-06-07 2020-12-09 Biotest AG Method and kit for testing potency of immunoglobulin compositions
CN113480622A (zh) * 2021-08-05 2021-10-08 江苏坤力生物制药有限责任公司 一种重组肺炎球菌溶血素制备及纯化的方法
JP2024533373A (ja) 2021-09-09 2024-09-12 アフィニバックス、インコーポレイテッド 多価肺炎球菌ワクチン

Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1990006951A1 (en) 1988-12-16 1990-06-28 De Staat Der Nederlanden Vertegenwoordigd Door De Minister Van Welzijn, Volksgezondheid En Cultuur Pneumolysin mutants and pneumococcal vaccines made therefrom
US6019982A (en) 1994-08-26 2000-02-01 The Administrators Of The Tulane Educational Fund Mutant enterotoxin effective as a non-toxic oral adjuvant
US20020187154A1 (en) 1993-12-22 2002-12-12 Rino Rappuoli Nontoxic mucosal adjuvant
US20030072774A1 (en) 1994-06-10 2003-04-17 Diane M. Gajewczyk Proteinaceous adjuvants
US6716432B1 (en) 1988-12-16 2004-04-06 James Cleland Paton Pneumolysin mutants and pneumococcal vaccines made therefrom
US6764686B2 (en) 1997-07-21 2004-07-20 Baxter International Inc. Modified immunogenic pneumolysin compositions as vaccines
WO2005108580A1 (en) * 2004-05-07 2005-11-17 Lea-Ann Kirkham Mutant pneumolysin proteins
WO2008022302A2 (en) * 2006-08-17 2008-02-21 The Uab Research Foundation Immunogenic pcpa polypeptides and uses thereof
US20100143399A1 (en) 2007-01-04 2010-06-10 Glaxosmithkline Biologicals Process for manufacturing vaccines
US8128939B2 (en) 2007-04-13 2012-03-06 The Board Of Regents Of The University Of Oklahoma Mutants of cholesterol-dependent cytolysins and uses thereof

Family Cites Families (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5364773A (en) 1991-03-07 1994-11-15 Virogenetics Corporation Genetically engineered vaccine strain
US5833975A (en) 1989-03-08 1998-11-10 Virogenetics Corporation Canarypox virus expressing cytokine and/or tumor-associated antigen DNA sequence
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
JP3011939B2 (ja) 1987-03-02 2000-02-21 ホワイトヘツド・インスチチユート・フオー・バイオメデイカル・リサーチ 組換えミコバクテリア・ワクチン
US5504005A (en) 1987-03-02 1996-04-02 Albert Einstein College Of Medicine Of Yeshiva University Recombinant mycobacterial vaccine
GB8823869D0 (en) 1988-10-12 1988-11-16 Medical Res Council Production of antibodies
WO1990014363A1 (en) 1989-05-19 1990-11-29 Amgen Inc. Metalloproteinase inhibitor
CA2076753A1 (en) 1990-02-26 1991-08-27 Anthony J. Radford Shuttle plasmid for escherichia coli and mycobacteria
GB9015888D0 (en) 1990-07-19 1990-09-05 Smithkline Biolog Vectors
US5625126A (en) 1990-08-29 1997-04-29 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
US5633425A (en) 1990-08-29 1997-05-27 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5661016A (en) 1990-08-29 1997-08-26 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US5545806A (en) 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
KR100272077B1 (ko) 1990-08-29 2000-11-15 젠팜인터내셔날,인코포레이티드 이종 항체를 생산할 수 있는 전이유전자를 가진 인간이외의 동물
AU672359B2 (en) 1991-03-07 1996-10-03 Virogenetics Corporation Genetically engineered vaccine strain
AU2185592A (en) 1991-06-06 1993-01-08 Med Immune, Inc. Induction of ctl responses to foreign antigens expressed in mycobacteria
US5990091A (en) 1997-03-12 1999-11-23 Virogenetics Corporation Vectors having enhanced expression, and methods of making and uses thereof
US7111252B1 (en) 1999-09-22 2006-09-19 Harris Scott C Enhancing touch and feel on the internet
US6530482B1 (en) 2000-04-26 2003-03-11 Michael D. Wiseman Tandem shale shaker
WO2007144647A2 (en) * 2006-06-15 2007-12-21 Timothy John Mitchell Adjuvant compositions

Patent Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1990006951A1 (en) 1988-12-16 1990-06-28 De Staat Der Nederlanden Vertegenwoordigd Door De Minister Van Welzijn, Volksgezondheid En Cultuur Pneumolysin mutants and pneumococcal vaccines made therefrom
US6716432B1 (en) 1988-12-16 2004-04-06 James Cleland Paton Pneumolysin mutants and pneumococcal vaccines made therefrom
US20020187154A1 (en) 1993-12-22 2002-12-12 Rino Rappuoli Nontoxic mucosal adjuvant
US20030072774A1 (en) 1994-06-10 2003-04-17 Diane M. Gajewczyk Proteinaceous adjuvants
US6019982A (en) 1994-08-26 2000-02-01 The Administrators Of The Tulane Educational Fund Mutant enterotoxin effective as a non-toxic oral adjuvant
US6764686B2 (en) 1997-07-21 2004-07-20 Baxter International Inc. Modified immunogenic pneumolysin compositions as vaccines
WO2005108580A1 (en) * 2004-05-07 2005-11-17 Lea-Ann Kirkham Mutant pneumolysin proteins
WO2008022302A2 (en) * 2006-08-17 2008-02-21 The Uab Research Foundation Immunogenic pcpa polypeptides and uses thereof
US20100227341A1 (en) 2006-08-17 2010-09-09 The Uab Research Foundation Diagnosing Pneumococcal Pneumonia
US20100143399A1 (en) 2007-01-04 2010-06-10 Glaxosmithkline Biologicals Process for manufacturing vaccines
US8128939B2 (en) 2007-04-13 2012-03-06 The Board Of Regents Of The University Of Oklahoma Mutants of cholesterol-dependent cytolysins and uses thereof

Non-Patent Citations (45)

* Cited by examiner, † Cited by third party
Title
Alexander, et al. Immunization of mice with pneumolysin toxoid confers a significant degree of protection against at least nine serotypes of Streptococcus pneumoniae. Infect. Immun. 62(12):5683-8 (1994).
Benton, et al.. A pneumolysin-negative mutant of Streptococcus pneumoniae causes chronic bacteremia rather than acute sepsis in mice. Infect. Immun. 63(2):448-55 (1995).
Bergeron, et al. Cytokine kinetics and other host factors in response to pneumococcal pulmonary infection in mice. Infect. Immun. 66(3):912-22 (1998).
Berry, et al. Comparative virulence of Streptococcus pneumoniae strains with insertion-duplication, point, and deletion mutations in the pneumolysin gene. Infect. Immun. 67(2):981-5 (1999).
Berry, et al. Effect of defined point mutations in the pneumolysin gene on the virulence of Streptococcus pneumoniae. Infect. Immun. 63(5):1969-74 (1995).
Black, et al. In Plotkin, S. et al. eds. Vaccines, 5th Ed., WB Saunders, Chapter 23 (2008).
Braun, et al. Pneumolysin, a protein toxin of Streptococcus pneumoniae, induces nitric oxide production from macrophages. Infect. Immun. 67(8):3750-6 (1999).
Briles, et al. Immunizations with pneumococcal surface protein A and pneumolysin are protective against pneumonia in a murine model of pulmonary infection with Streptococcus pneumoniae. J. Infect. Dis. 188(3):339-48 (2003).
Butler, et al. Serotype Distribution of Streptococcus pneumoniae Infections among Preschool Children in the United States, 1978-1994 : Implications for Development of a Conjugate Vaccine. J. Infect. Dis. 171(4): 855-889 (1995).
Cockeran, et al. The role of pneumolysin in the pathogenesis of Streptococcus pneumoniae infection. Curr. Opin. Infect. Dis. 15(3):235-9 (2002).
De Los Toyos, et al. Functional analysis of pneumolysin by use of monoclonal antibodies. Infect. Immun. 64 (2):480-4 (1996).
Feldman, et al. Pneumolysin induces the salient histologic features of pneumococcal infection in the rat lung in vivo. Am. J. Respir. Cell. Mol. Biol. 5(5):416-23 (1991).
Fickl, et al. Pneumolysin-mediated activation of NFkappaB in human neutrophils is antagonized by docosahexaenoic acid. Clin. Exp. Immunol. 140(2):274-81 (2005).
Garcia, et al. Pneumococcal disease and vaccination in the Americas: an agenda for accelerated vaccine introduction. Rev. Panam. Salud. Publica. 19(5): 340-8 (2006).
Garcia-Suarez, et al. The role of pneumolysin in mediating lung damage in a lethal pneumococcal pneumonia murine model. Respir. Res. 8:3 (2007).
Genbank Accession No. M17717, 1987.
Hanage, et al. Invasiveness of Serotypes and Clones of Streptococcus pneumoniae among Children in Finland. Infect. Immun. 73(1): 431-5 (2005).
Huo, et al. Antibody response to pneumolysin and to pneumococcal capsular polysaccharide in healthy individuals and Streptococcus pneumoniae infected patients. Vaccine, 22(9-10):1157-61 (2004).
Jeffries, et al. Presence of Nonhemolytic Pneumolysin in Serotypes of Streptococcus pneumoniae Associated with Disease Outbreaks. J. Infect. Dis. 196(6): 936-44 (2007).
Kerr, et al. Role of inflammatory mediators in resistance and susceptibility to pneumococcal infection. Infect. Immun. 70(3):1547-57 (2002).
Kirkham, et al. Construction and immunological characterization of a novel nontoxic protective pneumolysin mutant for use in future pneumococcal vaccines. Infect. Immun. 74(1):586-93 (2006).
Korchev, et al. A conserved tryptophan in pneumolysin is a determinant of the characteristics of channels formed by pneumolysin in cells and planar lipid bilayers. Biochem J. 329 (Pt 3):571-7 (1998).
Lagos, R. Population-based surveillance for hospitalized and ambulatory pediatric invasive pneumococcal disease in Santiago, Chile. Ped. Infect. Dis. J. 21(12): 1115-23 (2002).
Lock, et al. Comparative efficacy of pneumococcal neuraminidase and pneumolysin as immunogens protective against Streptococcus pneumoniae. Microb. Pathog. 5(6):461-7 (1998).
Lu, et al. Protection against Pneumococcal colonization and fatal pneumonia by a trivalent conjugate of a fusion protein with the cell wall polysaccharide. Infect. Immun. 77(5):2076-83 (2009).
Malley, et al. Recognition of pneumolysin by Toll-like receptor 4 confers resistance to pneumococcal infection. PNAS USA, 100(4): 1966-71 (2003).
Marriot, et al. Pneumolysin: a double-edged sword during the host-pathogen interaction. Curr. Mol. Med. 8 (6):497-509 (2008).
Maus, et al. Pneumolysin-induced lung injury is independent of leukocyte trafficking into the alveolar space. J. Immunol. 173(2):1307-12 (2004).
Michelow, et al. Epidemiology and clinical characteristics of community-acquired pneumonia in hospitalized children. Pediatrics, 113(4):701-7 (2004).
Michon, et al. Multivalent pneumococcal capsular polysaccharide conjugate vaccines employing genetically detoxified pneumolysin as a carrier protein. Vaccine, 16(18):1732-41 (1998).
Mitchell, et al. Comparison of pneumolysin genes and proteins from Streptococcus pneumoniae types 1 and 2. Nuc. Acids Res. 18: 4010 (1990).
Nato, et al. Production and Characterization of Neutralizing and Nonneutralizing Monoclonal Antibodies against Listeriolysin O. Infect. Immun. 59(12): 4641-4646 (1991).
Obaro, S. Prospects for pneumococcal vaccination in African children. Acta Trop. 75(2):141-53 (2000).
O'Brien, et al. Burden of disease caused by Streptococcus pneumoniae in children younger than 5 years: global estimates. Lancet, 374(9693):893-902 (2009).
Ogunniyi, et al. Development of a vaccine against invasive pneumococcal disease based on combinations of virulence proteins of Streptococcus pneumoniae. Infect. Immun. 75(1):350-7 (2007).
Ogunniyi, et al. Immunization of mice with combinations of pneumococcal virulence proteins elicits enhanced protection against challenge with Streptococcus pneumoniae. Infect. Immun. 68(5):3028-33 (2000).
Ogunniyi, et al. Protection against Streptococcus pneumoniae elicited by immunization with pneumolysin and CbpA. Infect. Immun. 69(10):5997-6003 (2001).
Oloo, et al. Structure-guided antigen engineering yields pneumolysin mutants suitable for vaccination against pneumococcal disease. J. Biol. Chem. 286(14): 12133-12140 (2011).
Paton, et al. Effect of immunization with pneumolysin on survival time of mice challenged with Streptococcus pneumoniae. Infect. Immun. 40(2):548-52 (1983).
Paton, et al. Purification and immunogenicity of genetically obtained pneumolysin toxoids and their conjugation to Streptococcus pneumoniae type 19F polysaccharide. Infect. Immun. 59(7):2297-304 (1991).
Rijneveld, et al. Roles of interleukin-6 and macrophage inflammatory protein-2 in pneumolysin-induced lung inflammation in mice. J. Infect. Dis. 185(1):123-6 (2002).
Rubins, et al. Pneumolysin in pneumococcal adherence and colonization. Microb. Pathog. 25(6):337-42 (1998).
Rubins, et al. Toxicity of pneumolysin to pulmonary alveolar epithelial cells. Infect. Immun. 61(4):1352-8 (1993).
Siber, et al. Pneumococcal Vaccines: The Impact of Conjugate Vaccine. JAMA, 301(6): 673-4 (2008).
Walker, et al. Molecular Cloning, Characterization, and Complete Nucleotide Sequence of the Gene for Pneumolysin, the Sulfhydryl-Activated Toxin of Streptococcus pneumoniae. Infect. Immun. 55(5): 1184-1187 (1987).

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10562941B2 (en) 2014-11-21 2020-02-18 The Board Of Regents Of The University Of Oklahoma Pneumolysin mutants and methods of use thereof
US10973908B1 (en) 2020-05-14 2021-04-13 David Gordon Bermudes Expression of SARS-CoV-2 spike protein receptor binding domain in attenuated salmonella as a vaccine
US11406702B1 (en) 2020-05-14 2022-08-09 David Gordon Bermudes Expression of SARS-CoV-2 spike protein receptor binding domain in attenuated Salmonella as a vaccine

Also Published As

Publication number Publication date
CA2748149C (en) 2018-03-06
US20110287046A1 (en) 2011-11-24
KR20110128800A (ko) 2011-11-30
MX2011006922A (es) 2012-06-12
IL213734A (he) 2017-09-28
EP2376526A4 (en) 2013-02-27
US8951531B2 (en) 2015-02-10
BRPI0923649A2 (pt) 2016-01-19
IL229526A0 (he) 2013-12-31
CN106008679A (zh) 2016-10-12
EP2376526A1 (en) 2011-10-19
CA2748149A1 (en) 2010-07-01
IL213734A0 (en) 2011-07-31
US9546199B2 (en) 2017-01-17
AU2009329782A1 (en) 2011-08-04
AU2009329782B2 (en) 2016-01-28
CN102712680A (zh) 2012-10-03
CN102712680B (zh) 2016-06-22
WO2010071986A1 (en) 2010-07-01
KR101741426B1 (ko) 2017-05-30
US20140341942A1 (en) 2014-11-20
US20150376242A1 (en) 2015-12-31
ZA201104627B (en) 2012-05-25

Similar Documents

Publication Publication Date Title
US9546199B2 (en) Modified Streptococcus pneumoniae pneumolysin (PLY) polypeptides
EP2183366B1 (en) Immunogenic polypeptides and monoclonal antibodies
US9366673B2 (en) Porphyromonas gingivalis polypeptides
KR20070122458A (ko) 표재성 스트렙토코커스 뉴모니아 폴리펩티드
AU2008204471B2 (en) Protective proteins of S. agalactiae, combinations thereof and methods of using the same
AU2013204613A1 (en) Modified Streptococcus pneumonia pneumolysin (PLY) polypeptides
AU2016254447A1 (en) Modified Streptococcus pneumonia pneumolysin (PLY) polypeptides
Class et al. Patent application title: Immunogenic Polypeptides and Monoclonal Antibodies Inventors: Martina Ochs (Toronto, CA) Roger Brooks (Toronto, CA) Robert Charlebois (Toronto, CA) Jeremy Yethon (Milton, CA)
US20130184200A1 (en) Immunogenic Polypeptides and Monoclonal Antibodies
CA2634911A1 (en) Neisseria meningitidis vaccines and their use

Legal Events

Date Code Title Description
AS Assignment

Owner name: NETHERLANDS VACCINE INSTITUTE, NETHERLANDS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:SANOFI PASTEUR LIMITED;REEL/FRAME:027890/0457

Effective date: 20111104

Owner name: THE KINGDOM OF THE NETHERLANDS, REPRESENTED BY THE

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:NETHERLANDS VACCINE INSTITUTE;REEL/FRAME:027890/0690

Effective date: 20111216

Owner name: SANOFI PASTEUR LIMITED, CANADA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:OLOO, ELIUD;OOMEN, RAYMOND;OCHS-ONOLEMHEMHEN, MARTINA;AND OTHERS;REEL/FRAME:027890/0373

Effective date: 20100318

STCF Information on status: patent grant

Free format text: PATENTED CASE

FEPP Fee payment procedure

Free format text: MAINTENANCE FEE REMINDER MAILED (ORIGINAL EVENT CODE: REM.)

FEPP Fee payment procedure

Free format text: SURCHARGE FOR LATE PAYMENT, LARGE ENTITY (ORIGINAL EVENT CODE: M1554)

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 4TH YEAR, LARGE ENTITY (ORIGINAL EVENT CODE: M1551)

Year of fee payment: 4

FEPP Fee payment procedure

Free format text: MAINTENANCE FEE REMINDER MAILED (ORIGINAL EVENT CODE: REM.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

LAPS Lapse for failure to pay maintenance fees

Free format text: PATENT EXPIRED FOR FAILURE TO PAY MAINTENANCE FEES (ORIGINAL EVENT CODE: EXP.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

STCH Information on status: patent discontinuation

Free format text: PATENT EXPIRED DUE TO NONPAYMENT OF MAINTENANCE FEES UNDER 37 CFR 1.362

FP Lapsed due to failure to pay maintenance fee

Effective date: 20220624